51
|
Abstract
Stem cells (SCs) maintain tissue homeostasis and repair wounds. Despite marked variation in tissue architecture and regenerative demands, SCs often follow similar paradigms in communicating with their microenvironmental "niche" to transition between quiescent and regenerative states. Here we use skin epithelium and skeletal muscle-among the most highly-stressed tissues in our body-to highlight similarities and differences in niche constituents and how SCs mediate natural tissue rejuvenation and perform regenerative acts prompted by injuries. We discuss how these communication networks break down during aging and how understanding tissue SCs has led to major advances in regenerative medicine.
Collapse
Affiliation(s)
- Elaine Fuchs
- Robin Chemers Neustein Laboratory of Mammalian Cell Biology and Development, Howard Hughes Medical Institute, The Rockefeller University, New York, NY 10065, USA.
| | - Helen M Blau
- Baxter Foundation Laboratory for Stem Cell Biology, Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, CA 94305, USA.
| |
Collapse
|
52
|
Schutt C, Hallmann A, Hachim S, Klockner I, Valussi M, Atzberger A, Graumann J, Braun T, Boettger T. Linc-MYH configures INO80 to regulate muscle stem cell numbers and skeletal muscle hypertrophy. EMBO J 2020; 39:e105098. [PMID: 32960481 PMCID: PMC7667881 DOI: 10.15252/embj.2020105098] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2020] [Revised: 08/20/2020] [Accepted: 08/24/2020] [Indexed: 12/16/2022] Open
Abstract
Chromatin remodeling complexes have functions in transcriptional regulation and chromosome maintenance, but it is mostly unknown how the function of these normally ubiquitous complexes is specified in the cellular context. Here, we describe that the evolutionary conserved long non‐coding RNA linc‐MYH regulates the composition of the INO80 chromatin remodeler complex in muscle stem cells and prevents interaction with WDR5 and the transcription factor YY1. Linc‐MYH acts as a selective molecular switch in trans that governs the pro‐proliferative function of the ubiquitous INO80 complex but does not affect its role in maintaining genomic stability. The molecular switch is essential for restricting generation of quiescent MuSCs and proliferation of myoblasts in homeostasis and regeneration. Since linc‐MYH is expressed in proliferating myoblasts but not in quiescent MuSCs, we reason that the extent of myoblast proliferation has decisive effects on the size of the quiescent MuSC pool.
Collapse
Affiliation(s)
- Christian Schutt
- Department of Cardiac Development and Remodelling, Max Planck Institute for Heart- and Lung Research, Bad Nauheim, Germany
| | - Alix Hallmann
- Department of Cardiac Development and Remodelling, Max Planck Institute for Heart- and Lung Research, Bad Nauheim, Germany
| | - Salma Hachim
- Department of Cardiac Development and Remodelling, Max Planck Institute for Heart- and Lung Research, Bad Nauheim, Germany
| | - Ina Klockner
- Department of Cardiac Development and Remodelling, Max Planck Institute for Heart- and Lung Research, Bad Nauheim, Germany
| | - Melissa Valussi
- Department of Cardiac Development and Remodelling, Max Planck Institute for Heart- and Lung Research, Bad Nauheim, Germany
| | - Ann Atzberger
- Max Planck Institute for Heart- and Lung Research, FACS Service Group, Bad Nauheim, Germany
| | - Johannes Graumann
- Max Planck Institute for Heart- and Lung Research, Mass Spectrometry Service Group, Bad Nauheim, Germany
| | - Thomas Braun
- Department of Cardiac Development and Remodelling, Max Planck Institute for Heart- and Lung Research, Bad Nauheim, Germany
| | - Thomas Boettger
- Department of Cardiac Development and Remodelling, Max Planck Institute for Heart- and Lung Research, Bad Nauheim, Germany
| |
Collapse
|
53
|
Yablonka-Reuveni Z, Lepper C. New Insight into a Classic Stem Cell: the Satellite Cell may Communicate with the Muscle Fiber via Extracellular Vesicles-A Perspective on "Fusion-Independent Satellite Cell Communication to Muscle Fibers During Load-Induced Hypertrophy". FUNCTION (OXFORD, ENGLAND) 2020; 1:zqaa015. [PMID: 35330641 PMCID: PMC8788824 DOI: 10.1093/function/zqaa015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 08/26/2020] [Accepted: 09/01/2020] [Indexed: 01/06/2023]
Affiliation(s)
- Zipora Yablonka-Reuveni
- Department of Biological Structure, School of Medicine, University of Washington, 1959 NE Pacific Street, Box 357420, Seattle, WA 98195, USA,Address correspondence to Z.Y.-R. (e-mail: ), C.L. (e-mail: )
| | - Christoph Lepper
- Department of Physiology & Cell Biology, College of Medicine, The Ohio State University, 1645 Neil Ave., Columbus, OH 43210, USA,Address correspondence to Z.Y.-R. (e-mail: ), C.L. (e-mail: )
| |
Collapse
|
54
|
Eye alignment changes caused by sustained GDNF treatment of an extraocular muscle in infant non-human primates. Sci Rep 2020; 10:11927. [PMID: 32681083 PMCID: PMC7368047 DOI: 10.1038/s41598-020-68743-3] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2020] [Accepted: 06/04/2020] [Indexed: 12/16/2022] Open
Abstract
The ability of sustained treatment of a single extraocular muscle with glial cell line-derived neurotrophic factor (GDNF) to produce a strabismus in infant non-human primates was tested. Six infant non-human primates received a pellet containing GDNF, releasing 2 µg/day for 90 days, on one medial rectus muscle. Eye alignment was assessed up to 6 months. Five of the six animals showed a slow decrease in eye misalignment from the significant exotropia present at birth, ending with approximately 10° of exotropia. Controls became orthotropic. Misalignment averaged 8° three months after treatment ended. After sustained GDNF treatment, few changes were seen in mean myofiber cross-sectional areas compared to age-matched naïve controls. Neuromuscular junction number was unaltered in the medial rectus muscles, but were significantly reduced in the untreated lateral recti. Neuromuscular junctions on slow fibers became multiply innervated after this sustained GDNF treatment. Pitx2-positive cells significantly decreased in treated and contralateral medial rectus muscles. Our study suggests that balanced GDNF signaling plays a role in normal development and maintenance of orthotropia. Sustained GDNF treatment of one medial rectus muscle resulted in a measurable misalignment largely maintained 3 months after treatment ended. Structural changes suggest mechanisms for producing an imbalance in muscle function.
Collapse
|
55
|
Masschelein E, D'Hulst G, Zvick J, Hinte L, Soro-Arnaiz I, Gorski T, von Meyenn F, Bar-Nur O, De Bock K. Exercise promotes satellite cell contribution to myofibers in a load-dependent manner. Skelet Muscle 2020; 10:21. [PMID: 32646489 PMCID: PMC7346400 DOI: 10.1186/s13395-020-00237-2] [Citation(s) in RCA: 52] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2020] [Accepted: 06/15/2020] [Indexed: 01/07/2023] Open
Abstract
BACKGROUND Satellite cells (SCs) are required for muscle repair following injury and are involved in muscle remodeling upon muscular contractions. Exercise stimulates SC accumulation and myonuclear accretion. To what extent exercise training at different mechanical loads drive SC contribution to myonuclei however is unknown. RESULTS By performing SC fate tracing experiments, we show that 8 weeks of voluntary wheel running increased SC contribution to myofibers in mouse plantar flexor muscles in a load-dependent, but fiber type-independent manner. Increased SC fusion however was not exclusively linked to muscle hypertrophy as wheel running without external load substantially increased SC fusion in the absence of fiber hypertrophy. Due to nuclear propagation, nuclear fluorescent fate tracing mouse models were inadequate to quantify SC contribution to myonuclei. Ultimately, by performing fate tracing at the DNA level, we show that SC contribution mirrors myonuclear accretion during exercise. CONCLUSIONS Collectively, mechanical load during exercise independently promotes SC contribution to existing myofibers. Also, due to propagation of nuclear fluorescent reporter proteins, our data warrant caution for the use of existing reporter mouse models for the quantitative evaluation of satellite cell contribution to myonuclei.
Collapse
Affiliation(s)
- Evi Masschelein
- Department Health Sciences and Technology, Laboratory of Exercise and Health, Swiss Federal Institute of Technology (ETH) Zurich, Zurich, Switzerland
| | - Gommaar D'Hulst
- Department Health Sciences and Technology, Laboratory of Exercise and Health, Swiss Federal Institute of Technology (ETH) Zurich, Zurich, Switzerland
| | - Joel Zvick
- Department Health Sciences and Technology, Laboratory of Regenerative and Movement Biology, Swiss Federal Institute of Technology (ETH) Zurich, Zurich, Switzerland
| | - Laura Hinte
- Department Health Sciences and Technology, Laboratory of Nutrition and Metabolic Epigenetics, Swiss Federal Institute of Technology (ETH) Zurich, Zurich, Switzerland
| | - Inés Soro-Arnaiz
- Department Health Sciences and Technology, Laboratory of Exercise and Health, Swiss Federal Institute of Technology (ETH) Zurich, Zurich, Switzerland
| | - Tatiane Gorski
- Department Health Sciences and Technology, Laboratory of Exercise and Health, Swiss Federal Institute of Technology (ETH) Zurich, Zurich, Switzerland
| | - Ferdinand von Meyenn
- Department Health Sciences and Technology, Laboratory of Nutrition and Metabolic Epigenetics, Swiss Federal Institute of Technology (ETH) Zurich, Zurich, Switzerland
| | - Ori Bar-Nur
- Department Health Sciences and Technology, Laboratory of Regenerative and Movement Biology, Swiss Federal Institute of Technology (ETH) Zurich, Zurich, Switzerland
| | - Katrien De Bock
- Department Health Sciences and Technology, Laboratory of Exercise and Health, Swiss Federal Institute of Technology (ETH) Zurich, Zurich, Switzerland.
| |
Collapse
|
56
|
Murach KA, Vechetti IJ, Van Pelt DW, Crow SE, Dungan CM, Figueiredo VC, Kosmac K, Fu X, Richards CI, Fry CS, McCarthy JJ, Peterson CA. Fusion-Independent Satellite Cell Communication to Muscle Fibers During Load-Induced Hypertrophy. FUNCTION 2020; 1:zqaa009. [PMID: 32864621 PMCID: PMC7448100 DOI: 10.1093/function/zqaa009] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2020] [Revised: 06/29/2020] [Accepted: 06/29/2020] [Indexed: 01/06/2023] Open
Abstract
The "canonical" function of Pax7+ muscle stem cells (satellite cells) during hypertrophic growth of adult muscle fibers is myonuclear donation via fusion to support increased transcriptional output. In recent years, however, emerging evidence suggests that satellite cells play an important secretory role in promoting load-mediated growth. Utilizing genetically modified mouse models of delayed satellite cell fusion and in vivo extracellular vesicle (EV) tracking, we provide evidence for satellite cell communication to muscle fibers during hypertrophy. Myogenic progenitor cell-EV-mediated communication to myotubes in vitro influences extracellular matrix (ECM)-related gene expression, which is congruent with in vivo overload experiments involving satellite cell depletion, as well as in silico analyses. Satellite cell-derived EVs can transfer a Cre-induced, cytoplasmic-localized fluorescent reporter to muscle cells as well as microRNAs that regulate ECM genes such as matrix metalloproteinase 9 (Mmp9), which may facilitate growth. Delayed satellite cell fusion did not limit long-term load-induced muscle hypertrophy indicating that early fusion-independent communication from satellite cells to muscle fibers is an underappreciated aspect of satellite cell biology. We cannot exclude the possibility that satellite cell-mediated myonuclear accretion is necessary to maintain prolonged growth, specifically in the later phases of adaptation, but these data collectively highlight how EV delivery from satellite cells can directly contribute to mechanical load-induced muscle fiber hypertrophy, independent of cell fusion to the fiber.
Collapse
Affiliation(s)
- Kevin A Murach
- The Center for Muscle Biology, University of Kentucky, Lexington, KY 40536, USA
- Department of Physical Therapy, College of Health Sciences, University of Kentucky, Lexington, KY 40536, USA
| | - Ivan J Vechetti
- The Center for Muscle Biology, University of Kentucky, Lexington, KY 40536, USA
- Department of Physiology, College of Medicine, University of Kentucky, Lexington, KY 40536, USA
| | - Douglas W Van Pelt
- The Center for Muscle Biology, University of Kentucky, Lexington, KY 40536, USA
- Department of Physical Therapy, College of Health Sciences, University of Kentucky, Lexington, KY 40536, USA
| | - Samuel E Crow
- The Center for Muscle Biology, University of Kentucky, Lexington, KY 40536, USA
- Department of Physical Therapy, College of Health Sciences, University of Kentucky, Lexington, KY 40536, USA
| | - Cory M Dungan
- The Center for Muscle Biology, University of Kentucky, Lexington, KY 40536, USA
- Department of Physical Therapy, College of Health Sciences, University of Kentucky, Lexington, KY 40536, USA
| | - Vandre C Figueiredo
- The Center for Muscle Biology, University of Kentucky, Lexington, KY 40536, USA
- Department of Physical Therapy, College of Health Sciences, University of Kentucky, Lexington, KY 40536, USA
| | - Kate Kosmac
- The Center for Muscle Biology, University of Kentucky, Lexington, KY 40536, USA
- Department of Physical Therapy, College of Health Sciences, University of Kentucky, Lexington, KY 40536, USA
| | - Xu Fu
- Department of Chemistry, College of Arts and Sciences, University of Kentucky, Lexington, KY 40536, USA
| | - Christopher I Richards
- Department of Chemistry, College of Arts and Sciences, University of Kentucky, Lexington, KY 40536, USA
| | - Christopher S Fry
- The Center for Muscle Biology, University of Kentucky, Lexington, KY 40536, USA
- Department of Athletic Training and Clinical Nutrition, College of Health Sciences, University of Kentucky, Lexington, KY 40536, USA
| | - John J McCarthy
- The Center for Muscle Biology, University of Kentucky, Lexington, KY 40536, USA
- Department of Physiology, College of Medicine, University of Kentucky, Lexington, KY 40536, USA
| | - Charlotte A Peterson
- The Center for Muscle Biology, University of Kentucky, Lexington, KY 40536, USA
- Department of Physical Therapy, College of Health Sciences, University of Kentucky, Lexington, KY 40536, USA
- Department of Physiology, College of Medicine, University of Kentucky, Lexington, KY 40536, USA
| |
Collapse
|
57
|
Arneson-Wissink PC, Hogan KA, Ducharme AM, Samani A, Jatoi A, Doles JD. The wasting-associated metabolite succinate disrupts myogenesis and impairs skeletal muscle regeneration. JCSM RAPID COMMUNICATIONS 2020; 3:56-69. [PMID: 32905522 PMCID: PMC7470228 DOI: 10.1002/rco2.14] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/02/2023]
Abstract
BACKGROUND Muscle wasting is a debilitating co-morbidity affecting most advanced cancer patients. Alongside enhanced muscle catabolism, defects in muscle repair/regeneration contribute to cancer-associated wasting. Among the factors implicated in suppression of muscle regeneration are cytokines that interfere with myogenic signal transduction pathways. Less understood is how other cancer/wasting-associated cues, such as metabolites, contribute to muscle dysfunction. This study investigates how the metabolite succinate affects myogenesis and muscle regeneration. METHODS We leveraged an established ectopic metabolite treatment (cell permeable dimethyl-succinate) strategy to evaluate the ability of intracellular succinate elevation to 1) affect myoblast homeostasis (proliferation, apoptosis), 2) disrupt protein dynamics and induce wasting-associated atrophy, and 3) modulate in vitro myogenesis. In vivo succinate supplementation experiments (2% succinate, 1% sucrose vehicle) were used to corroborate and extend in vitro observations. Metabolic profiling and functional metabolic studies were then performed to investigate the impact of succinate elevation on mitochondria function. RESULTS We found that in vitro succinate supplementation elevated intracellular succinate about 2-fold, and did not have an impact on proliferation or apoptosis of C2C12 myoblasts. Elevated succinate had minor effects on protein homeostasis (~25% decrease in protein synthesis assessed by OPP staining), and no significant effect on myotube atrophy. Succinate elevation interfered with in vitro myoblast differentiation, characterized by significant decreases in late markers of myogenesis and fewer nuclei per myosin heavy chain positive structure (assessed by immunofluorescence staining). While mice orally administered succinate did not exhibit changes in overall body composition or whole muscle weights, these mice displayed smaller muscle myofiber diameters (~6% decrease in the mean of non-linear regression curves fit to the histograms of minimum feret diameter distribution), which was exacerbated when muscle regeneration was induced with barium chloride injury. Significant decreases in the mean of non-linear regression curves fit to the histograms of minimum feret diameter distributions were observed 7 days and 28 days post injury. Elevated numbers of myogenin positive cells (3-fold increase) supportive of the differentiation defects observed in vitro were observed 28 days post injury. Metabolic profiling and functional metabolic assessment of myoblasts revealed that succinate elevation caused both widespread metabolic changes and significantly lowered maximal cellular respiration (~35% decrease). CONCLUSIONS This study broadens the repertoire of wasting-associated factors that can directly modulate muscle progenitor cell function and strengthens the hypothesis that metabolic derangements are significant contributors to impaired muscle regeneration, an important aspect of cancer-associated muscle wasting.
Collapse
Affiliation(s)
- Paige C Arneson-Wissink
- Department of Biochemistry and Molecular Biology, Mayo
Clinic, Rochester, Minnesota, 55905 USA
| | - Kelly A Hogan
- Department of Biochemistry and Molecular Biology, Mayo
Clinic, Rochester, Minnesota, 55905 USA
| | - Alexandra M Ducharme
- Department of Biochemistry and Molecular Biology, Mayo
Clinic, Rochester, Minnesota, 55905 USA
| | - Adrienne Samani
- Department of Biochemistry and Molecular Biology, Mayo
Clinic, Rochester, Minnesota, 55905 USA
| | - Aminah Jatoi
- Department of Oncology, Mayo Clinic, Rochester,
Minnesota
| | - Jason D Doles
- Department of Biochemistry and Molecular Biology, Mayo
Clinic, Rochester, Minnesota, 55905 USA
- Corresponding Author: Jason D Doles, Department of
Biochemistry and Molecular Biology, Mayo Clinic, 200 First St SW, Guggenheim
16-11A1, Rochester, MN 55905, Tel: (507) 284-9372, Fax: (507) 284-3383,
| |
Collapse
|
58
|
Al Tanoury Z, Rao J, Tassy O, Gobert B, Gapon S, Garnier JM, Wagner E, Hick A, Hall A, Gussoni E, Pourquié O. Differentiation of the human PAX7-positive myogenic precursors/satellite cell lineage in vitro. Development 2020; 147:dev187344. [PMID: 32541004 PMCID: PMC7328153 DOI: 10.1242/dev.187344] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2019] [Accepted: 05/29/2020] [Indexed: 12/12/2022]
Abstract
Satellite cells (SC) are muscle stem cells that can regenerate adult muscles upon injury. Most SC originate from PAX7+ myogenic precursors set aside during development. Although myogenesis has been studied in mouse and chicken embryos, little is known about human muscle development. Here, we report the generation of human induced pluripotent stem cell (iPSC) reporter lines in which fluorescent proteins have been introduced into the PAX7 and MYOG loci. We use single cell RNA sequencing to analyze the developmental trajectory of the iPSC-derived PAX7+ myogenic precursors. We show that the PAX7+ cells generated in culture can produce myofibers and self-renew in vitro and in vivo Together, we demonstrate that cells exhibiting characteristics of human fetal satellite cells can be produced in vitro from iPSC, opening interesting avenues for muscular dystrophy cell therapy. This work provides significant insights into the development of the human myogenic lineage.
Collapse
Affiliation(s)
- Ziad Al Tanoury
- Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), Department of Development and Stem Cells, CNRS (UMR 7104), Inserm U964, Université de Strasbourg, 67404, Illkirch Graffenstaden, France
- Department of Pathology, Brigham and Women's Hospital, 60 Fenwood Road, Boston, MA 02115, USA
- Department of Genetics, Harvard Medical School, 60 Fenwood Road, Boston, MA 02115, USA
| | - Jyoti Rao
- Department of Pathology, Brigham and Women's Hospital, 60 Fenwood Road, Boston, MA 02115, USA
- Department of Genetics, Harvard Medical School, 60 Fenwood Road, Boston, MA 02115, USA
| | - Olivier Tassy
- Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), Department of Development and Stem Cells, CNRS (UMR 7104), Inserm U964, Université de Strasbourg, 67404, Illkirch Graffenstaden, France
| | - Bénédicte Gobert
- Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), Department of Development and Stem Cells, CNRS (UMR 7104), Inserm U964, Université de Strasbourg, 67404, Illkirch Graffenstaden, France
- Anagenesis Biotechnologies, Parc d'innovation - BioParc 3, 850 Boulevard Sébastien Brandt, 67400 Illkirch Graffenstaden, France
| | - Svetlana Gapon
- Department of Pathology, Brigham and Women's Hospital, 60 Fenwood Road, Boston, MA 02115, USA
| | - Jean-Marie Garnier
- Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), Department of Development and Stem Cells, CNRS (UMR 7104), Inserm U964, Université de Strasbourg, 67404, Illkirch Graffenstaden, France
| | - Erica Wagner
- Department of Pathology, Brigham and Women's Hospital, 60 Fenwood Road, Boston, MA 02115, USA
| | - Aurore Hick
- Anagenesis Biotechnologies, Parc d'innovation - BioParc 3, 850 Boulevard Sébastien Brandt, 67400 Illkirch Graffenstaden, France
| | - Arielle Hall
- Division of Genetics and Genomics, Boston Children's Hospital, 3 Blackfan Circle, CLS, Boston, MA 15021, USA
| | - Emanuela Gussoni
- Division of Genetics and Genomics, Boston Children's Hospital, 3 Blackfan Circle, CLS, Boston, MA 15021, USA
| | - Olivier Pourquié
- Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), Department of Development and Stem Cells, CNRS (UMR 7104), Inserm U964, Université de Strasbourg, 67404, Illkirch Graffenstaden, France
- Department of Pathology, Brigham and Women's Hospital, 60 Fenwood Road, Boston, MA 02115, USA
- Department of Genetics, Harvard Medical School, 60 Fenwood Road, Boston, MA 02115, USA
- Harvard Stem Cell Institute, Harvard University, Cambridge, MA 02138, USA
| |
Collapse
|
59
|
Abstract
Changes in muscle stem cell (MuSC) function during aging have been assessed using various in vivo and ex vivo systems. However, changes in clonal complexity within the aged MuSC pool are relatively understudied. Although the dissection of stem cell heterogeneity has greatly benefited from several technological advancements, including single cell sequencing, these methods preclude longitudinal measures of individual stem cell behavior. Instead, multicolor labeling systems enable lineage tracing with single cell resolution. Here, we describe a method of inducibly labeling MuSCs with the Brainbow-2.1 multicolor lineage tracing reporter in vivo to track individual MuSC fate and assess clonal complexity in the overall MuSC pool throughout the mouse lifespan.
Collapse
|
60
|
In Vivo Genome Engineering for the Treatment of Muscular Dystrophies. CURRENT STEM CELL REPORTS 2020. [DOI: 10.1007/s40778-020-00173-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
|
61
|
Lee C, Agha O, Liu M, Davies M, Bertoy L, Kim HT, Liu X, Feeley BT. Rotator Cuff Fibro-Adipogenic Progenitors Demonstrate Highest Concentration, Proliferative Capacity, and Adipogenic Potential Across Muscle Groups. J Orthop Res 2020; 38:1113-1121. [PMID: 31799698 PMCID: PMC9262119 DOI: 10.1002/jor.24550] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/26/2019] [Accepted: 11/30/2019] [Indexed: 02/04/2023]
Abstract
Fatty infiltration (FI) of rotator cuff (RC) muscles is common in patients with RC tears. Studies have demonstrated that fibro-adipogenic progenitors (FAPs), a population of resident muscle stem cells, are the main contributors of FI, which adversely affects muscle quality and RC repair success. Although FI is common in RC injuries, it is not frequently reported after other musculotendinous injuries. Additionally, studies have shown the development of different pathology patterns across muscle groups suggestive of intrinsic differences in cellular composition and behavior. This study evaluates FAP distribution and differentiation properties across anatomic locations in mice. Muscles from seven different anatomic locations were harvested from PDGFRα-eGFP FAP reporter mice. FAPs were quantified using histology and FACS sorting with BD Aria II with CD31- /CD45- /Integrinα7- /Sca-1+ and PDGFRα reporter signal (n = 3 per muscle). The cells were analyzed for adipogenesis using immunocytochemistry and for proliferation properties with Brdu-Ki67 staining. In a separate group of mice, RC and tibialis anterior muscles received glycerol injection and were harvested after 2 weeks for FI quantification (n = 4). One-way analysis of variance was used for statistical comparisons among groups, with significance at p < 0.05. FAPs from the RC, masseter, and paraspinal muscles were more numerous and demonstrated greater proliferative capacity and adipogenic potency than those from the tibialis anterior and gastrocnemius. The RC demonstrated significantly greater levels of FI than the tibialis anterior after glycerol-injection injury. Clinical Significance: This study suggests differences in FAP distribution and differentiation characteristics may account for the propensity to develop FI in RC tears as compared with other musculotendinous injuries. © 2019 Orthopaedic Research Society. Published by Wiley Periodicals, Inc. J Orthop Res 38:1113-1121, 2020.
Collapse
Affiliation(s)
- Carlin Lee
- San Francisco Veteran Affairs Health Care System, San Francisco, California, 94158,Department of Orthopaedic Surgery, University of California San Francisco, San Francisco, California, 94158
| | - Obiajulu Agha
- San Francisco Veteran Affairs Health Care System, San Francisco, California, 94158,Department of Orthopaedic Surgery, University of California San Francisco, San Francisco, California, 94158
| | - Mengyao Liu
- San Francisco Veteran Affairs Health Care System, San Francisco, California, 94158,Department of Orthopaedic Surgery, University of California San Francisco, San Francisco, California, 94158
| | - Michael Davies
- San Francisco Veteran Affairs Health Care System, San Francisco, California, 94158,Department of Orthopaedic Surgery, University of California San Francisco, San Francisco, California, 94158
| | - Lauren Bertoy
- San Francisco Veteran Affairs Health Care System, San Francisco, California, 94158,Department of Orthopaedic Surgery, University of California San Francisco, San Francisco, California, 94158
| | - Hubert T. Kim
- San Francisco Veteran Affairs Health Care System, San Francisco, California, 94158,Department of Orthopaedic Surgery, University of California San Francisco, San Francisco, California, 94158
| | - Xuhui Liu
- San Francisco Veteran Affairs Health Care System, San Francisco, California, 94158,Department of Orthopaedic Surgery, University of California San Francisco, San Francisco, California, 94158
| | - Brian T. Feeley
- San Francisco Veteran Affairs Health Care System, San Francisco, California, 94158,Department of Orthopaedic Surgery, University of California San Francisco, San Francisco, California, 94158
| |
Collapse
|
62
|
Englund DA, Murach KA, Dungan CM, Figueiredo VC, Vechetti IJ, Dupont-Versteegden EE, McCarthy JJ, Peterson CA. Depletion of resident muscle stem cells negatively impacts running volume, physical function, and muscle fiber hypertrophy in response to lifelong physical activity. Am J Physiol Cell Physiol 2020; 318:C1178-C1188. [PMID: 32320286 DOI: 10.1152/ajpcell.00090.2020] [Citation(s) in RCA: 64] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
To date, studies that have aimed to investigate the role of satellite cells during adult skeletal muscle adaptation and hypertrophy have utilized a nontranslational stimulus and/or have been performed over a relatively short time frame. Although it has been shown that satellite cell depletion throughout adulthood does not drive skeletal muscle loss in sedentary mice, it remains unknown how satellite cells participate in skeletal muscle adaptation to long-term physical activity. The current study was designed to determine whether reduced satellite cell content throughout adulthood would influence the transcriptome-wide response to physical activity and diminish the adaptive response of skeletal muscle. We administered vehicle or tamoxifen to adult Pax7-diphtheria toxin A (DTA) mice to deplete satellite cells and assigned them to sedentary or wheel-running conditions for 13 mo. Satellite cell depletion throughout adulthood reduced balance and coordination, overall running volume, and the size of muscle proprioceptors (spindle fibers). Furthermore, satellite cell participation was necessary for optimal muscle fiber hypertrophy but not adaptations in fiber type distribution in response to lifelong physical activity. Transcriptome-wide analysis of the plantaris and soleus revealed that satellite cell function is muscle type specific; satellite cell-dependent myonuclear accretion was apparent in oxidative muscles, whereas initiation of G protein-coupled receptor (GPCR) signaling in the glycolytic plantaris may require satellite cells to induce optimal adaptations to long-term physical activity. These findings suggest that satellite cells play a role in preserving physical function during aging and influence muscle adaptation during sustained periods of physical activity.
Collapse
Affiliation(s)
- Davis A Englund
- Department of Physical Therapy, College of Health Sciences, University of Kentucky, Lexington, Kentucky.,Center for Muscle Biology, University of Kentucky, Lexington, Kentucky
| | - Kevin A Murach
- Department of Physical Therapy, College of Health Sciences, University of Kentucky, Lexington, Kentucky.,Center for Muscle Biology, University of Kentucky, Lexington, Kentucky
| | - Cory M Dungan
- Department of Physical Therapy, College of Health Sciences, University of Kentucky, Lexington, Kentucky.,Center for Muscle Biology, University of Kentucky, Lexington, Kentucky
| | - Vandré C Figueiredo
- Department of Physical Therapy, College of Health Sciences, University of Kentucky, Lexington, Kentucky.,Center for Muscle Biology, University of Kentucky, Lexington, Kentucky
| | - Ivan J Vechetti
- Center for Muscle Biology, University of Kentucky, Lexington, Kentucky.,Department of Physiology, College of Medicine, University of Kentucky, Lexington, Kentucky
| | - Esther E Dupont-Versteegden
- Department of Physical Therapy, College of Health Sciences, University of Kentucky, Lexington, Kentucky.,Center for Muscle Biology, University of Kentucky, Lexington, Kentucky
| | - John J McCarthy
- Center for Muscle Biology, University of Kentucky, Lexington, Kentucky.,Department of Physiology, College of Medicine, University of Kentucky, Lexington, Kentucky
| | - Charlotte A Peterson
- Department of Physical Therapy, College of Health Sciences, University of Kentucky, Lexington, Kentucky.,Center for Muscle Biology, University of Kentucky, Lexington, Kentucky
| |
Collapse
|
63
|
de Morree A, Klein JDD, Gan Q, Farup J, Urtasun A, Kanugovi A, Bilen B, van Velthoven CTJ, Quarta M, Rando TA. Alternative polyadenylation of Pax3 controls muscle stem cell fate and muscle function. Science 2020; 366:734-738. [PMID: 31699935 DOI: 10.1126/science.aax1694] [Citation(s) in RCA: 58] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2019] [Accepted: 09/26/2019] [Indexed: 12/16/2022]
Abstract
Adult stem cells are essential for tissue homeostasis. In skeletal muscle, muscle stem cells (MuSCs) reside in a quiescent state, but little is known about the mechanisms that control homeostatic turnover. Here we show that, in mice, the variation in MuSC activation rate among different muscles (for example, limb versus diaphragm muscles) is determined by the levels of the transcription factor Pax3. We further show that Pax3 levels are controlled by alternative polyadenylation of its transcript, which is regulated by the small nucleolar RNA U1. Isoforms of the Pax3 messenger RNA that differ in their 3' untranslated regions are differentially susceptible to regulation by microRNA miR206, which results in varying levels of the Pax3 protein in vivo. These findings highlight a previously unrecognized mechanism of the homeostatic regulation of stem cell fate by multiple RNA species.
Collapse
Affiliation(s)
- Antoine de Morree
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA, USA. .,Paul F. Glenn Center for the Biology of Aging, Stanford University School of Medicine, Stanford, CA, USA
| | - Julian D D Klein
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA, USA.,Paul F. Glenn Center for the Biology of Aging, Stanford University School of Medicine, Stanford, CA, USA
| | - Qiang Gan
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA, USA.,Paul F. Glenn Center for the Biology of Aging, Stanford University School of Medicine, Stanford, CA, USA
| | - Jean Farup
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA, USA.,Paul F. Glenn Center for the Biology of Aging, Stanford University School of Medicine, Stanford, CA, USA.,Departments of Clinical Medicine and Biomedicine, Research Laboratory for Biochemical Pathology, Aarhus University, Aarhus, Denmark
| | - Andoni Urtasun
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA, USA.,Paul F. Glenn Center for the Biology of Aging, Stanford University School of Medicine, Stanford, CA, USA
| | - Abhijnya Kanugovi
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA, USA.,Paul F. Glenn Center for the Biology of Aging, Stanford University School of Medicine, Stanford, CA, USA
| | - Biter Bilen
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA, USA.,Paul F. Glenn Center for the Biology of Aging, Stanford University School of Medicine, Stanford, CA, USA
| | - Cindy T J van Velthoven
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA, USA.,Paul F. Glenn Center for the Biology of Aging, Stanford University School of Medicine, Stanford, CA, USA
| | - Marco Quarta
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA, USA.,Paul F. Glenn Center for the Biology of Aging, Stanford University School of Medicine, Stanford, CA, USA.,Center for Tissue Regeneration, Repair, and Restoration, Veterans Affairs Palo Alto Health Care System, Palo Alto, CA, USA
| | - Thomas A Rando
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA, USA. .,Paul F. Glenn Center for the Biology of Aging, Stanford University School of Medicine, Stanford, CA, USA.,Center for Tissue Regeneration, Repair, and Restoration, Veterans Affairs Palo Alto Health Care System, Palo Alto, CA, USA
| |
Collapse
|
64
|
Carrero-Rojas G, Benítez-Temiño B, Pastor AM, Davis López de Carrizosa MA. Muscle Progenitors Derived from Extraocular Muscles Express Higher Levels of Neurotrophins and their Receptors than other Cranial and Limb Muscles. Cells 2020; 9:cells9030747. [PMID: 32197508 PMCID: PMC7140653 DOI: 10.3390/cells9030747] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2020] [Revised: 03/02/2020] [Accepted: 03/17/2020] [Indexed: 01/19/2023] Open
Abstract
Extraocular muscles (EOMs) show resistance to muscle dystrophies and sarcopenia. It has been recently demonstrated that they are endowed with different types of myogenic cells, all of which present an outstanding regenerative potential. Neurotrophins are important modulators of myogenic regeneration and act promoting myoblast proliferation, enhancing myogenic fusion rates and protecting myotubes from inflammatory stimuli. Here, we adapted the pre-plate cell isolation technique to obtain myogenic progenitors from the rat EOMs, and quantified their in vitro expression of neurotrophins and their receptors by RT–qPCR and immunohistochemistry, respectively. The results were compared with the expression on progenitors isolated from buccinator, tongue and limb muscles. Our quantitative analysis of brain-derived neurotrophic factor (BDNF), nerve growth factor (NGF) and neurotrophin-3 (NT-3) transcripts showed, for the first time, that EOMs-derived cells express more of these factors and that they expressed TrkA, but not TrkB and TrkC receptors. On the contrary, the immunofluorescence analysis demonstrated high expression of p75NTR on all myogenic progenitors, with the EOMs-derived cells showing higher expression. Taken together, these results suggest that the intrinsic trophic differences between EOMs-derived myogenic progenitors and their counterparts from other muscles could explain why those cells show higher proliferative and fusion rates, as well as better regenerative properties.
Collapse
|
65
|
Yousuf Y, Datu A, Barnes B, Amini-Nik S, Jeschke MG. Metformin alleviates muscle wasting post-thermal injury by increasing Pax7-positive muscle progenitor cells. Stem Cell Res Ther 2020; 11:18. [PMID: 31915055 PMCID: PMC6950874 DOI: 10.1186/s13287-019-1480-x] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2019] [Revised: 10/14/2019] [Accepted: 10/31/2019] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Profound skeletal muscle wasting and weakness is common after severe burn and persists for years after injury contributing to morbidity and mortality of burn patients. Currently, no ideal treatment exists to inhibit muscle catabolism. Metformin is an anti-diabetic agent that manages hyperglycemia but has also been shown to have a beneficial effect on stem cells after injury. We hypothesize that metformin administration will increase protein synthesis in the skeletal muscle by increasing the proliferation of muscle progenitor cells, thus mitigating muscle atrophy post-burn injury. METHODS To determine whether metformin can attenuate muscle catabolism following burn injury, we utilized a 30% total burn surface area (TBSA) full-thickness scald burn in mice and compared burn injuries with and without metformin treatment. We examined the gastrocnemius muscle at 7 and 14 days post-burn injury. RESULTS At 7 days, burn injury significantly reduced myofiber cross-sectional area (CSA) compared to sham, p < 0.05. Metformin treatment significantly attenuated muscle catabolism and preserved muscle CSA at the sham size. To investigate metformin's effect on satellite cells (muscle progenitors), we examined changes in Pax7, a transcription factor regulating the proliferation of muscle progenitors. Burned animals treated with metformin had a significant increase in Pax7 protein level and the number of Pax7-positive cells at 7 days post-burn, p < 0.05. Moreover, through BrdU proliferation assay, we show that metformin treatment increased the proliferation of satellite cells at 7 days post-burn injury, p < 0.05. CONCLUSION In summary, metformin's various metabolic effects and its modulation of stem cells make it an attractive alternative to mitigate burn-induced muscle wasting while also managing hyperglycemia.
Collapse
Affiliation(s)
- Yusef Yousuf
- Sunnybrook Research Institute, 2075 Bayview Ave., Rm. D704, Toronto, ON, M4N 3M5, Canada
| | - Andrea Datu
- Sunnybrook Research Institute, 2075 Bayview Ave., Rm. D704, Toronto, ON, M4N 3M5, Canada
| | - Ben Barnes
- Sunnybrook Research Institute, 2075 Bayview Ave., Rm. D704, Toronto, ON, M4N 3M5, Canada
| | - Saeid Amini-Nik
- Sunnybrook Research Institute, 2075 Bayview Ave., Rm. D704, Toronto, ON, M4N 3M5, Canada. .,Laboratory in Medicine and Pathobiology, University of Toronto, Toronto, Canada. .,Division of Plastic Surgery, Department of Surgery, University of Toronto, Toronto, Canada.
| | - Marc G Jeschke
- Sunnybrook Research Institute, 2075 Bayview Ave., Rm. D704, Toronto, ON, M4N 3M5, Canada. .,Division of Plastic Surgery, Department of Surgery, University of Toronto, Toronto, Canada. .,Ross Tilley Burn Centre, Sunnybrook Health Sciences Centre, Toronto, Canada. .,Department of Immunology, University of Toronto, Toronto, Canada.
| |
Collapse
|
66
|
Muñoz‐Cánoves P, Neves J, Sousa‐Victor P. Understanding muscle regenerative decline with aging: new approaches to bring back youthfulness to aged stem cells. FEBS J 2020; 287:406-416. [DOI: 10.1111/febs.15182] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2019] [Revised: 11/21/2019] [Accepted: 12/17/2019] [Indexed: 12/17/2022]
Affiliation(s)
- Pura Muñoz‐Cánoves
- Department of Experimental and Health Sciences CIBERNED ICREA University Pompeu Fabra (UPF) Barcelona Spain
- Spanish National Cardiovascular Research Center Madrid Spain
| | - Joana Neves
- Instituto de Medicina Molecular (iMM) Faculdade de Medicina Universidade de Lisboa Portugal
| | - Pedro Sousa‐Victor
- Instituto de Medicina Molecular (iMM) Faculdade de Medicina Universidade de Lisboa Portugal
| |
Collapse
|
67
|
Rimer M. Extracellular signal-regulated kinases 1 and 2 regulate neuromuscular junction and myofiber phenotypes in mammalian skeletal muscle. Neurosci Lett 2019; 715:134671. [PMID: 31805372 DOI: 10.1016/j.neulet.2019.134671] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2019] [Revised: 11/27/2019] [Accepted: 11/30/2019] [Indexed: 02/06/2023]
Abstract
The neuromuscular junction is the synapse between a motor neuron of the spinal cord and a skeletal muscle fiber in the periphery. Reciprocal interactions between these excitable cells, and between them and others cell types present within the muscle tissue, shape the development, homeostasis and plasticity of skeletal muscle. An important aim in the field is to understand the molecular mechanisms underlying these cellular interactions, which include identifying the nature of the signals and receptors involved but also of the downstream intracellular signaling cascades elicited by them. This review focuses on work that shows that skeletal muscle fiber-derived extracellular signal-regulated kinases 1 and 2 (ERK1/2), ubiquitous and prototypical intracellular mitogen-activated protein kinases, have modulatory roles in the maintenance of the neuromuscular synapse and in the acquisition and preservation of fiber type identity in skeletal muscle.
Collapse
Affiliation(s)
- Mendell Rimer
- Department of Neuroscience & Experimental Therapeutics, College of Medicine, Texas A&M Health Science Center and Texas A&M Institute for Neuroscience, Bryan, TX 77807 USA.
| |
Collapse
|
68
|
Medler S. Mixing it up: the biological significance of hybrid skeletal muscle fibers. ACTA ACUST UNITED AC 2019; 222:222/23/jeb200832. [PMID: 31784473 DOI: 10.1242/jeb.200832] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Skeletal muscle fibers are classified according to the myosin heavy chain (MHC) isoforms and other myofibrillar proteins expressed within these cells. In addition to 'pure' fibers expressing single MHC isoforms, many fibers are 'hybrids' that co-express two or more different isoforms of MHC or other myofibrillar proteins. Although hybrid fibers have been recognized by muscle biologists for more than three decades, uncertainty persists about their prevalence in normal muscles, their role in fiber-type transitions, and what they might tell us about fiber-type regulation at the cellular and molecular levels. This Review summarizes current knowledge on the relative abundance of hybrid fibers in a variety of muscles from different species. Data from more than 150 muscles from 39 species demonstrate that hybrid fibers are common, frequently representing 25% or more of the fibers in normal muscles. Hybrid fibers appear to have two main roles: (1) they function as intermediates during the fiber-type transitions associated with skeletal muscle development, adaptation to exercise and aging; and (2) they provide a functional continuum of fiber phenotypes, as they possess physiological properties that are intermediate to those of pure fiber types. One aspect of hybrid fibers that is not widely recognized is that fiber-type asymmetries - such as dramatic differences in the MHC composition along the length of single fibers - appear to be a common aspect of many fibers. The final section of this Review examines the possible role of differential activities of nuclei in different myonuclear domains in establishing fiber-type asymmetries.
Collapse
Affiliation(s)
- Scott Medler
- Biology Department, State University of New York at Fredonia, Fredonia, NY 14063, USA
| |
Collapse
|
69
|
Ruparelia AA, Ratnayake D, Currie PD. Stem cells in skeletal muscle growth and regeneration in amniotes and teleosts: Emerging themes. WILEY INTERDISCIPLINARY REVIEWS-DEVELOPMENTAL BIOLOGY 2019; 9:e365. [PMID: 31743958 DOI: 10.1002/wdev.365] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/06/2019] [Revised: 09/22/2019] [Accepted: 10/03/2019] [Indexed: 12/19/2022]
Abstract
Skeletal muscle is a contractile, postmitotic tissue that retains the capacity to grow and regenerate throughout life in amniotes and teleost. Both muscle growth and regeneration are regulated by obligate tissue resident muscle stem cells. Given that considerable knowledge exists on the myogenic process, recent studies have focused on examining the molecular markers of muscle stem cells, and on the intrinsic and extrinsic signals regulating their function. From this, two themes emerge: firstly, muscle stem cells display remarkable heterogeneity not only with regards to their gene expression profile, but also with respect to their behavior and function; and secondly, the stem cell niche is a critical regulator of muscle stem cell function during growth and regeneration. Here, we will address the current understanding of these emerging themes with emphasis on the distinct processes used by amniotes and teleost, and discuss the challenges and opportunities in the muscle growth and regeneration fields. This article is characterized under: Adult Stem Cells, Tissue Renewal, and Regeneration > Tissue Stem Cells and Niches Early Embryonic Development > Development to the Basic Body Plan Vertebrate Organogenesis > Musculoskeletal and Vascular.
Collapse
Affiliation(s)
- Avnika A Ruparelia
- Australian Regenerative Medicine Institute, Monash University, Melbourne, Victoria, Australia.,EMBL Australia, Monash University, Melbourne, Victoria, Australia
| | - Dhanushika Ratnayake
- Australian Regenerative Medicine Institute, Monash University, Melbourne, Victoria, Australia.,EMBL Australia, Monash University, Melbourne, Victoria, Australia
| | - Peter D Currie
- Australian Regenerative Medicine Institute, Monash University, Melbourne, Victoria, Australia.,EMBL Australia, Monash University, Melbourne, Victoria, Australia
| |
Collapse
|
70
|
A Three-Dimensional Culture Model of Reversibly Quiescent Myogenic Cells. Stem Cells Int 2019; 2019:7548160. [PMID: 31827532 PMCID: PMC6885280 DOI: 10.1155/2019/7548160] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2019] [Revised: 09/24/2019] [Accepted: 10/03/2019] [Indexed: 12/13/2022] Open
Abstract
Satellite cells (SC) are the stem cells of skeletal muscles. They are quiescent in adult animals but resume proliferation to allow muscle hypertrophy or regeneration after injury. The mechanisms balancing quiescence, self-renewal, and differentiation of SC are difficult to analyze in vivo owing to their complexity and in vitro because the staminal character of SC is lost when they are removed from the niche and is not adequately reproduced in the culture models currently available. To overcome these difficulties, we set up a culture model of the myogenic C2C12 cell line in suspension. When C2C12 cells are cultured in suspension, they enter a state of quiescence and form three-dimensional aggregates (myospheres) that produce the extracellular matrix and express markers of quiescent SC. In the initial phase of culture, a portion of the cells fuses in syncytia and abandons the myospheres. The remaining cells are mononucleated and quiescent but resume proliferation and differentiation when plated in a monolayer. The notch pathway controls the quiescent state of the cells as shown by the fact that its inhibition leads to the resumption of differentiation. Within this context, notch3 appears to play a central role in the activity of this pathway since the expression of notch1 declines soon after aggregation. In summary, the culture model of C2C12 in suspension may be used to study the cellular interactions of muscle stem cells and the pathways controlling SC quiescence entrance and maintenance.
Collapse
|
71
|
Affiliation(s)
- Haibin Xi
- Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, Department of Microbiology, Immunology, and Molecular Genetics, David Geffen School of Medicine at University of California-Los Angeles, Los Angeles, CA 90095, USA
| | - April Pyle
- Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, Department of Microbiology, Immunology, and Molecular Genetics, David Geffen School of Medicine at University of California-Los Angeles, Los Angeles, CA 90095, USA.
| |
Collapse
|
72
|
Bahri OA, Naldaiz-Gastesi N, Kennedy DC, Wheatley AM, Izeta A, McCullagh KJA. The panniculus carnosus muscle: A novel model of striated muscle regeneration that exhibits sex differences in the mdx mouse. Sci Rep 2019; 9:15964. [PMID: 31685850 PMCID: PMC6828975 DOI: 10.1038/s41598-019-52071-2] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2019] [Accepted: 10/10/2019] [Indexed: 01/12/2023] Open
Abstract
The dermal striated muscle panniculus carnosus (PC), prevalent in lower mammals with remnants in humans, is highly regenerative, and whose function is purported to be linked to defence and shivering thermogenesis. Given the heterogeneity of responses of different muscles to disease, we set out to characterize the PC in wild-type and muscular dystrophic mdx mice. The mouse PC contained mainly fast-twitch type IIB myofibers showing body wide distribution. The PC exemplified heterogeneity in myofiber sizes and a prevalence of central nucleated fibres (CNFs), hallmarks of regeneration, in wild-type and mdx muscles, which increased with age. PC myofibers were hypertrophic in mdx compared to wild-type mice. Sexual dimorphism was apparent with a two-fold increase in CNFs in PC from male versus female mdx mice. To evaluate myogenic potential, PC muscle progenitors were isolated from 8-week old wild-type and mdx mice, grown and differentiated for 7-days. Myogenic profiling of PC-derived myocytes suggested that male mdx satellite cells (SCs) were more myogenic than female counterparts, independent of SC density in PC muscles. Muscle regenerative differences in the PC were associated with alterations in expression of calcium handling regulatory proteins. These studies highlight unique aspects of the PC muscle and its potential as a model to study mechanisms of striated muscle regeneration in health and disease.
Collapse
MESH Headings
- Animals
- Biomarkers
- Calcium-Binding Proteins/metabolism
- Cell Differentiation
- Dermis/metabolism
- Dermis/pathology
- Disease Models, Animal
- Female
- Immunohistochemistry
- Male
- Mice
- Mice, Inbred mdx
- Muscle Development
- Muscle Fibers, Skeletal/metabolism
- Muscle Fibers, Skeletal/pathology
- Muscle, Striated/pathology
- Muscle, Striated/physiology
- Muscular Dystrophy, Duchenne/metabolism
- Muscular Dystrophy, Duchenne/pathology
- Regeneration
- Satellite Cells, Skeletal Muscle/cytology
- Satellite Cells, Skeletal Muscle/metabolism
- Sex Factors
- Stem Cells
Collapse
Affiliation(s)
- Ola A Bahri
- Department of Physiology, School of Medicine, Human Biology Building, National University of Ireland Galway, Galway, H91 W5P7, Ireland
- Regenerative Medicine Institute, School of Medicine, National University of Ireland Galway, Galway, Ireland
| | | | - Donna C Kennedy
- Department of Physiology, School of Medicine, Human Biology Building, National University of Ireland Galway, Galway, H91 W5P7, Ireland
| | - Antony M Wheatley
- Department of Physiology, School of Medicine, Human Biology Building, National University of Ireland Galway, Galway, H91 W5P7, Ireland
| | - Ander Izeta
- Biodonostia Health Research Institute, San Sebastian, Spain
| | - Karl J A McCullagh
- Department of Physiology, School of Medicine, Human Biology Building, National University of Ireland Galway, Galway, H91 W5P7, Ireland.
- Regenerative Medicine Institute, School of Medicine, National University of Ireland Galway, Galway, Ireland.
| |
Collapse
|
73
|
Patel K, Simbi B, Ritvos O, Vaiyapuri S, Dhoot GK. Dysregulated cell signalling and reduced satellite cell potential in ageing muscle. Exp Cell Res 2019; 385:111685. [PMID: 31647919 DOI: 10.1016/j.yexcr.2019.111685] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2019] [Revised: 10/16/2019] [Accepted: 10/19/2019] [Indexed: 01/16/2023]
Abstract
Aberrant activation of signalling pathways has been postulated to promote age related changes in skeletal muscle. Cell signalling activation requires not only the expression of ligands and receptors but also an appropriate environment that facilitates their interaction. Here we first examined the expression of SULF1/SULF2 and members of RTK (receptor tyrosine kinase) and the Wnt family in skeletal muscle of normal and a mouse model of accelerated ageing. We show that SULF1/SULF2 and these signalling components, a feature of early muscle development are barely detectable in early postnatal muscle. Real time qPCR and immunocytochemical analysis showed gradual but progressive up-regulation of SULF1/SULF2 and RTK/Wnt proteins not only in the activated satellite cells but also on muscle fibres that gradually increased with age. Satellite cells on isolated muscle fibres showed spontaneous in vivo satellite cell activation and progressive reduction in proliferative potential and responsiveness to HGF (hepatocyte growth factor) and dysregulated myogenic differentiation with age. Finally, we show that SULF1/SULF2 and RTK/Wnt signalling components are expressed in progeric mouse muscles at earlier stage but their expression is attenuated by an intervention that promotes muscle repair and growth.
Collapse
Affiliation(s)
- Ketan Patel
- School of Biological Sciences, University of Reading, Reading, UK
| | - Biggy Simbi
- Department of Comparative Biomedical Sciences, Royal Veterinary College, University of London, UK
| | - Olli Ritvos
- Department of Bacteriology and Immunology, University of Helsinki, Helsinki, Finland
| | | | - Gurtej K Dhoot
- Department of Comparative Biomedical Sciences, Royal Veterinary College, University of London, UK.
| |
Collapse
|
74
|
Sema3a-Nrp1 Signaling Mediates Fast-Twitch Myofiber Specificity of Tw2 + Cells. Dev Cell 2019; 51:89-98.e4. [PMID: 31474563 DOI: 10.1016/j.devcel.2019.08.002] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2019] [Revised: 07/03/2019] [Accepted: 08/02/2019] [Indexed: 12/11/2022]
Abstract
We previously identified a unique population of interstitial muscle progenitors, marked by expression of the Twist2 transcription factor, which fuses specifically to type IIb/x fast-twitch myofibers. Tw2+ progenitors are distinct from satellite cells, a muscle progenitor that expresses Pax7 and contributes to all myofiber types. Through RNA sequencing and immunofluorescence, we identify the membrane receptor, Nrp1, as a marker of Tw2+ cells but not Pax7+ cells. We also found that Sema3a, a chemorepellent ligand for Nrp1, is expressed by type I and IIa myofibers but not IIb myofibers. Using stripe migration assays, chimeric cell-cell fusion assays, and a Sema3a transgenic mouse model, we identify Sema3a-Nrp1 signaling as a major mechanism for Tw2+ cell fiber-type specificity. Our findings reveal an extracellular signaling mechanism whereby a cell-surface receptor for a chemorepellent confers specificity of intercellular fusion of a specific muscle progenitor with its target tissue.
Collapse
|
75
|
Ato S, Kido K, Sato K, Fujita S. Type 2 diabetes causes skeletal muscle atrophy but does not impair resistance training-mediated myonuclear accretion and muscle mass gain in rats. Exp Physiol 2019; 104:1518-1531. [PMID: 31328833 PMCID: PMC6790689 DOI: 10.1113/ep087585] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2019] [Accepted: 07/19/2019] [Indexed: 12/12/2022]
Abstract
New Findings What is the central question of this study? Type 2 diabetes mellitus (T2DM) causes skeletal muscle atrophy; does it affect resistance training (RT)‐mediated molecular adaptations and subsequent muscle hypertrophy? What is the main finding and its importance? Although skeletal muscle mass and regulation were not preserved under conditions of T2DM, the response of RT‐induced skeletal muscle hypertrophy was not impaired in T2DM rat skeletal muscle. These findings suggest that the capacity of RT‐mediated muscle mass gain is not diminished in the T2DM condition.
Abstract Type 2 diabetes mellitus (T2DM) is known to cause skeletal muscle atrophy. However, it is not known whether T2DM affects resistance training (RT)‐mediated molecular adaptations and subsequent muscle hypertrophy. Therefore, we investigated the effect of T2DM on response of skeletal muscle hypertrophy to chronic RT using a rat resistance exercise mimetic model. T2DM and healthy control rats were subjected to 18 bouts (3 times per week) of chronic RT on unilateral lower legs. RT significantly increased gastrocnemius muscle mass and myonuclei in both T2DM and healthy control rats to the same extent, even though T2DM caused muscle atrophy in the resting condition. Further, T2DM significantly reduced mechanistic target of rapamycin complex 1 (mTORC1) activity (phosphorylation of p70S6KThr389 and 4E‐BP1Thr37/46) to insulin stimulation and the number of myonuclei in the untrained basal condition, but RT‐mediated adaptations were not affected by T2DM. These findings suggested that although the skeletal muscle mass and regulation were not preserved under basal conditions of T2DM, the response of RT‐induced skeletal muscle hypertrophy was not impaired in T2DM rat skeletal muscle.
Collapse
Affiliation(s)
- Satoru Ato
- Graduate School of Sport and Health Science, Ritsumeikan University, Kusatsu, Japan
| | - Kohei Kido
- Graduate School of Sport and Health Science, Ritsumeikan University, Kusatsu, Japan
| | - Koji Sato
- Faculty of Human Development, Kobe University, Kobe, Japan
| | - Satoshi Fujita
- Graduate School of Sport and Health Science, Ritsumeikan University, Kusatsu, Japan
| |
Collapse
|
76
|
Gordon SJV, Fenker DE, Vest KE, Padilla-Benavides T. Manganese influx and expression of ZIP8 is essential in primary myoblasts and contributes to activation of SOD2. Metallomics 2019; 11:1140-1153. [PMID: 31086870 PMCID: PMC6584035 DOI: 10.1039/c8mt00348c] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Trace elements such as copper (Cu), zinc (Zn), iron (Fe), and manganese (Mn) function as enzyme cofactors and second messengers in cell signaling. Trace elements are emerging as key regulators of differentiation and development of mammalian tissues including blood, brain, and skeletal muscle. We previously reported an influx of Cu and dynamic expression of metal transporters during differentiation of skeletal muscle cells. Here, we demonstrate that during differentiation of skeletal myoblasts an increase of Mn, Fe and Zn also occurs. Interestingly the Mn increase is concomitant with increased Mn-dependent SOD2 levels. To better understand the Mn import pathway in skeletal muscle cells, we probed the functional relevance of the closely related proteins ZIP8 and ZIP14, which are implicated in Zn, Mn, and Fe transport. Partial depletion of ZIP8 severely impaired growth of myoblasts and led to cell death under differentiation conditions, indicating that ZIP8-mediated metal transport is essential in skeletal muscle cells. Moreover, knockdown of Zip8 impaired activity of the Mn-dependent SOD2. Growth defects were partially rescued only by Mn supplementation to the medium, suggesting additional functions for ZIP8 in the skeletal muscle lineage. Restoring wild type Zip8 into the knockdown cells rescued the proliferation and differentiation phenotypes. On the other hand, knockdown of Zip14, had only a mild effect on myotube size, consistent with a role for ZIP14 in muscle hypertrophy. Simultaneous knockdown of both Zip8 and Zip14 further impaired differentiation and led cell death. This is the first report on the functional relevance of two members of the ZIP family of metal transporters in the skeletal muscle lineage, and further supports the paradigm that trace metal transporters are important modulators of mammalian tissue development.
Collapse
Affiliation(s)
- Shellaina J. V. Gordon
- Department of Biochemistry and Molecular Pharmacology,
University of Massachusetts Medical School, 394 Plantation St., Worcester, MA,
01605, USA
| | - Daniel E. Fenker
- Department of Molecular Genetics, Biochemistry &
Microbiology, University of Cincinnati School of Medicine, 231 Albert Sabin Way,
Cincinnati, OH, 45267, USA
| | - Katherine E. Vest
- Department of Molecular Genetics, Biochemistry &
Microbiology, University of Cincinnati School of Medicine, 231 Albert Sabin Way,
Cincinnati, OH, 45267, USA
| | - Teresita Padilla-Benavides
- Department of Biochemistry and Molecular Pharmacology,
University of Massachusetts Medical School, 394 Plantation St., Worcester, MA,
01605, USA
| |
Collapse
|
77
|
Muscle development and regeneration controlled by AUF1-mediated stage-specific degradation of fate-determining checkpoint mRNAs. Proc Natl Acad Sci U S A 2019; 116:11285-11290. [PMID: 31113881 DOI: 10.1073/pnas.1901165116] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
AUF1 promotes rapid decay of mRNAs containing 3' untranslated region (3'UTR) AU-rich elements (AREs). AUF1 depletion in mice accelerates muscle loss and causes limb girdle muscular dystrophy. Here, we demonstrate that the selective, targeted degradation by AUF1 of key muscle stem cell fate-determining checkpoint mRNAs regulates each stage of muscle development and regeneration by reprogramming each myogenic stage. Skeletal muscle stem (satellite) cell explants show that Auf1 transcription is activated with satellite cell activation by stem cell regulatory factor CTCF. AUF1 then targets checkpoint ARE-mRNAs for degradation, progressively reprogramming the transcriptome through each stage of myogenesis. Transition steps in myogenesis, from stem cell proliferation to differentiation to muscle fiber development, are each controlled by fate-determining checkpoint mRNAs, which, surprisingly, were found to be controlled in their expression by AUF1-targeted mRNA decay. Checkpoint mRNAs targeted by AUF1 include Twist1, decay of which promotes myoblast development; CyclinD1, decay of which blocks myoblast proliferation and initiates differentiation; and RGS5, decay of which activates Sonic Hedgehog (SHH) pathway-mediated differentiation of mature myotubes. AUF1 therefore orchestrates muscle stem cell proliferation, self-renewal, myoblast differentiation, and ultimately formation of muscle fibers through targeted, staged mRNA decay.
Collapse
|
78
|
Affiliation(s)
- Helen M Blau
- From the Baxter Laboratory for Stem Cell Biology, Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, CA (H.M.B.); and the Department of Medicine, Harvard Medical School, Boston (G.Q.D.)
| | - George Q Daley
- From the Baxter Laboratory for Stem Cell Biology, Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, CA (H.M.B.); and the Department of Medicine, Harvard Medical School, Boston (G.Q.D.)
| |
Collapse
|
79
|
PAX3 Confers Functional Heterogeneity in Skeletal Muscle Stem Cell Responses to Environmental Stress. Cell Stem Cell 2019; 24:958-973.e9. [PMID: 31006622 DOI: 10.1016/j.stem.2019.03.019] [Citation(s) in RCA: 60] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2018] [Revised: 02/04/2019] [Accepted: 03/22/2019] [Indexed: 11/22/2022]
Abstract
Muscle satellite cells (MuSCs) are the quiescent muscle stem cells required for adult skeletal muscle repair. The impact of environmental stress such as pollution on MuSC behavior remains unexplored. We evaluated the impact of 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) exposure, a ubiquitous and highly toxic pollutant, on MuSCs by combining in vivo mouse molecular genetic models with ex vivo studies. While all MuSCs express the transcription factor PAX7, we show that a subset also express PAX3 and exhibit resistance to environmental stress. Upon systemic TCDD treatment, PAX3-negative MuSCs display impaired survival, atypical activation, and sporadic differentiation through xenobiotic aryl hydrocarbon receptor signaling. We further show that PAX3-positive MuSCs become sensitized to environmental stress when PAX3 function is impaired and that PAX3-mediated induction of mTORC1 is required for protection. Our study, therefore, identifies a functional heterogeneity of MuSCs in response to environmental stress controlled by PAX3.
Collapse
|
80
|
High-Dimensional Single-Cell Cartography Reveals Novel Skeletal Muscle-Resident Cell Populations. Mol Cell 2019; 74:609-621.e6. [PMID: 30922843 DOI: 10.1016/j.molcel.2019.02.026] [Citation(s) in RCA: 249] [Impact Index Per Article: 41.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2018] [Revised: 12/13/2018] [Accepted: 02/14/2019] [Indexed: 12/12/2022]
Abstract
Adult tissue repair and regeneration require stem-progenitor cells that can self-renew and generate differentiated progeny. Skeletal muscle regenerative capacity relies on muscle satellite cells (MuSCs) and their interplay with different cell types within the niche. However, our understanding of skeletal muscle tissue cellular composition is limited. Here, using a combined approach of single-cell RNA sequencing and mass cytometry, we precisely mapped 10 different mononuclear cell types in adult mouse muscle. We also characterized gene signatures and determined key discriminating markers of each cell type. We identified two previously understudied cell populations in the interstitial compartment. One expresses the transcription factor scleraxis and generated tenocytes in vitro. The second expresses markers of smooth muscle and mesenchymal cells (SMMCs) and, while distinct from MuSCs, exhibited myogenic potential and promoted MuSC engraftment following transplantation. The blueprint presented here yields crucial insights into muscle-resident cell-type identities and can be exploited to study muscle diseases.
Collapse
|
81
|
Schubert MF, Noah AC, Bedi A, Gumucio JP, Mendias CL. Reduced Myogenic and Increased Adipogenic Differentiation Capacity of Rotator Cuff Muscle Stem Cells. J Bone Joint Surg Am 2019; 101:228-238. [PMID: 30730482 PMCID: PMC6791507 DOI: 10.2106/jbjs.18.00509] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
BACKGROUND Fat accumulation commonly occurs in chronically torn rotator cuff muscles, and increased fat within the rotator cuff is correlated with poor clinical outcomes. The extent of lipid deposition is particularly pronounced in injured rotator cuff muscles compared with other commonly injured muscles such as the gastrocnemius. Satellite cells, which are a tissue-resident muscle stem-cell population, can differentiate into fat cells. We hypothesized that satellite cells from the rotator cuff have greater intrinsic adipogenic differentiation potential than do gastrocnemius satellite cells, and this difference is due to variations in epigenetic imprinting between the cells. METHODS Satellite cells from gastrocnemius and rotator cuff muscles of mice were cultured in adipogenic media, and the capacity to differentiate into mature muscle cells and adipogenic cells was assessed (n ≥ 9 plates per muscle group). We also performed DNA methylation analysis of gastrocnemius and rotator cuff satellite cells to determine whether epigenetic differences were present between the 2 groups (n = 5 mice per group). RESULTS Compared with the gastrocnemius, satellite cells from the rotator cuff had a 23% reduction in myogenic differentiation and an 87% decrease in the expression of the differentiated muscle cell marker MRF4 (myogenic regulatory factor 4). With respect to adipogenesis, rotator cuff satellite cells had a 4.3-fold increase in adipogenesis, a 12-fold increase in the adipogenic transcription factor PPARγ (peroxisome proliferator-activated receptor gamma), and a 65-fold increase in the adipogenic marker FABP4 (fatty-acid binding protein 4). Epigenetic analysis identified 355 differentially methylated regions of DNA between rotator cuff and gastrocnemius satellite cells, and pathway enrichment analysis suggested that these regions were involved with lipid metabolism and adipogenesis. CONCLUSIONS Satellite cells from rotator cuff muscles have reduced myogenic and increased adipogenic differentiation potential compared with gastrocnemius muscles. There appears to be a cellular and genetic basis behind the generally poor rates of rotator cuff muscle healing. CLINICAL RELEVANCE The reduced myogenic and increased adipogenic capacity of rotator cuff satellite cells is consistent with the increased fat content and poor muscle healing rates often observed for chronically torn rotator cuff muscles. For patients undergoing rotator cuff repair, transplantation of autologous satellite cells from other muscles less prone to fatty infiltration may improve clinical outcomes.
Collapse
Affiliation(s)
- Manuel F. Schubert
- Departments of Orthopaedic Surgery (M.F.S., A.C.N., A.B, J.P.G, and C.L.M.) and Molecular and Integrative Physiology (A.C.N, J.P.G., and C.L.M.), University of Michigan Medical School, Ann Arbor, Michigan
| | - Andrew C. Noah
- Departments of Orthopaedic Surgery (M.F.S., A.C.N., A.B, J.P.G, and C.L.M.) and Molecular and Integrative Physiology (A.C.N, J.P.G., and C.L.M.), University of Michigan Medical School, Ann Arbor, Michigan
| | - Asheesh Bedi
- Departments of Orthopaedic Surgery (M.F.S., A.C.N., A.B, J.P.G, and C.L.M.) and Molecular and Integrative Physiology (A.C.N, J.P.G., and C.L.M.), University of Michigan Medical School, Ann Arbor, Michigan
| | - Jonathan P. Gumucio
- Departments of Orthopaedic Surgery (M.F.S., A.C.N., A.B, J.P.G, and C.L.M.) and Molecular and Integrative Physiology (A.C.N, J.P.G., and C.L.M.), University of Michigan Medical School, Ann Arbor, Michigan
| | - Christopher L. Mendias
- Departments of Orthopaedic Surgery (M.F.S., A.C.N., A.B, J.P.G, and C.L.M.) and Molecular and Integrative Physiology (A.C.N, J.P.G., and C.L.M.), University of Michigan Medical School, Ann Arbor, Michigan,Hospital for Special Surgery, New York, NY,Departments of Physiology and Biophysics and Orthopaedic Surgery, Weill Cornell Medical College, New York, NY
| |
Collapse
|
82
|
Franco I, Fernandez-Gonzalo R, Vrtačnik P, Lundberg TR, Eriksson M, Gustafsson T. Healthy skeletal muscle aging: The role of satellite cells, somatic mutations and exercise. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2019; 346:157-200. [DOI: 10.1016/bs.ircmb.2019.03.003] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|
83
|
Cho DS, Doles JD. Skeletal Muscle Progenitor Cell Heterogeneity. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2019; 1169:179-193. [PMID: 31487024 DOI: 10.1007/978-3-030-24108-7_9] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Tissue-specific stem cells contribute to adult tissue maintenance, repair, and regeneration. In skeletal muscle, many different mononuclear cell types are capable of giving rise to differentiated muscle. Of these tissue stem-like cells, satellite cells (SCs) are the most studied muscle stem cell population and are widely considered the main cellular source driving muscle repair and regeneration in adult tissue. Within the satellite cell pool, many distinct subpopulations exist, each exhibiting differential abilities to exit quiescence, expand, differentiate, and self-renew. In this chapter, we discuss the different stem cell types that can give rise to skeletal muscle tissue and then focus on satellite cell heterogeneity during the process of myogenesis/muscle regeneration. Finally, we highlight emerging opportunities to better characterize muscle stem cell heterogeneity, which will ultimately deepen our appreciation of stem cells in muscle development, repair/regeneration, aging, and disease.
Collapse
Affiliation(s)
- Dong Seong Cho
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN, USA
| | - Jason D Doles
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN, USA.
| |
Collapse
|
84
|
Gayraud-Morel B, Le Bouteiller M, Commere PH, Cohen-Tannoudji M, Tajbakhsh S. Notchless defines a stage-specific requirement for ribosome biogenesis during lineage progression in adult skeletal myogenesis. Development 2018; 145:145/23/dev162636. [PMID: 30478226 DOI: 10.1242/dev.162636] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2017] [Accepted: 10/02/2018] [Indexed: 11/20/2022]
Abstract
Cell fate decisions occur through the action of multiple factors, including signalling molecules and transcription factors. Recently, the regulation of translation has emerged as an important step for modulating cellular function and fate, as exemplified by ribosomes that play distinct roles in regulating cell behaviour. Notchless (Nle) is a conserved nuclear protein that is involved in a crucial step in ribosome biogenesis, and is required for the maintenance of adult haematopoietic and intestinal stem/progenitor cells. Here, we show that activated skeletal muscle satellite cells in conditional Nle mutant mice are arrested in proliferation; however, deletion of Nle in myofibres does not impair myogenesis. Furthermore, conditional deletion of Nle in satellite cells during homeostasis did not impact on their fate for up to 3 months. In contrast, loss of Nle function in primary myogenic cells blocked proliferation because of major defects in ribosome formation. Taken together, we show that muscle stem cells undergo a stage-specific regulation of ribosome biogenesis, thereby underscoring the importance of differential modulation of mRNA translation for controlling cell fate decisions.
Collapse
Affiliation(s)
- Barbara Gayraud-Morel
- Stem Cells and Development, Department of Developmental & Stem Cell Biology, Institut Pasteur, 25 rue du Dr Roux, 75015 Paris, France.,CNRS UMR 3738, Institut Pasteur, 25 rue du Dr Roux, 75015 Paris, France
| | - Marie Le Bouteiller
- CNRS UMR 3738, Institut Pasteur, 25 rue du Dr Roux, 75015 Paris, France.,Early Mammalian Development and Stem Cell Biology, Department of Developmental & Stem Cell Biology, Institut Pasteur, 25 rue du Dr Roux, 75015 Paris, France
| | - Pierre-Henri Commere
- Plateforme de Cytometrie, Institut Pasteur, 25 rue du Dr Roux, 75015 Paris, France
| | - Michel Cohen-Tannoudji
- CNRS UMR 3738, Institut Pasteur, 25 rue du Dr Roux, 75015 Paris, France.,Early Mammalian Development and Stem Cell Biology, Department of Developmental & Stem Cell Biology, Institut Pasteur, 25 rue du Dr Roux, 75015 Paris, France
| | - Shahragim Tajbakhsh
- Stem Cells and Development, Department of Developmental & Stem Cell Biology, Institut Pasteur, 25 rue du Dr Roux, 75015 Paris, France .,CNRS UMR 3738, Institut Pasteur, 25 rue du Dr Roux, 75015 Paris, France
| |
Collapse
|
85
|
Bachman JF, Klose A, Liu W, Paris ND, Blanc RS, Schmalz M, Knapp E, Chakkalakal JV. Prepubertal skeletal muscle growth requires Pax7-expressing satellite cell-derived myonuclear contribution. Development 2018; 145:dev.167197. [PMID: 30305290 PMCID: PMC6215399 DOI: 10.1242/dev.167197] [Citation(s) in RCA: 90] [Impact Index Per Article: 12.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2018] [Accepted: 09/18/2018] [Indexed: 12/14/2022]
Abstract
The functional role of Pax7-expressing satellite cells (SCs) in postnatal skeletal muscle development beyond weaning remains obscure. Therefore, the relevance of SCs during prepubertal growth, a period after weaning but prior to the onset of puberty, has not been examined. Here, we have characterized mouse skeletal muscle growth during prepuberty and found significant increases in myofiber cross-sectional area that correlated with SC-derived myonuclear number. Remarkably, genome-wide RNA-sequencing analysis established that post-weaning juvenile and early adolescent skeletal muscle have markedly different gene expression signatures. These distinctions are consistent with extensive skeletal muscle maturation during this essential, albeit brief, developmental phase. Indelible labeling of SCs with Pax7CreERT2/+; Rosa26nTnG/+ mice demonstrated SC-derived myonuclear contribution during prepuberty, with a substantial reduction at puberty onset. Prepubertal depletion of SCs in Pax7CreERT2/+; Rosa26DTA/+ mice reduced myofiber size and myonuclear number, and caused force generation deficits to a similar extent in both fast and slow-contracting muscles. Collectively, these data demonstrate SC-derived myonuclear accretion as a cellular mechanism that contributes to prepubertal hypertrophic skeletal muscle growth. Summary: Examination of gene expression and morphological changes in mouse skeletal muscle during prepuberty demonstrates that satellite cell-derived myonuclear accretion contributes to prepubertal hypertrophic skeletal muscle growth.
Collapse
Affiliation(s)
- John F Bachman
- Department of Pathology and Laboratory Medicine, Cell Biology of Disease Graduate Program, University of Rochester Medical Center, 601 Elmwood Ave, Rochester, NY 14642, USA.,Department of Pharmacology and Physiology, University of Rochester Medical Center, 601 Elmwood Ave Box 711, Rochester, NY 14642, USA
| | - Alanna Klose
- Department of Pharmacology and Physiology, University of Rochester Medical Center, 601 Elmwood Ave Box 711, Rochester, NY 14642, USA
| | - Wenxuan Liu
- Department of Pharmacology and Physiology, University of Rochester Medical Center, 601 Elmwood Ave Box 711, Rochester, NY 14642, USA .,Department of Biomedical Genetics, Genetics, Development, and Stem Cells Graduate Program, University of Rochester Medical Center, 601 Elmwood Ave Box 633, Rochester, NY 14642, USA
| | - Nicole D Paris
- Department of Pharmacology and Physiology, University of Rochester Medical Center, 601 Elmwood Ave Box 711, Rochester, NY 14642, USA
| | - Roméo S Blanc
- Department of Pharmacology and Physiology, University of Rochester Medical Center, 601 Elmwood Ave Box 711, Rochester, NY 14642, USA
| | - Melissa Schmalz
- Department of Pharmacology and Physiology, University of Rochester Medical Center, 601 Elmwood Ave Box 711, Rochester, NY 14642, USA
| | - Emma Knapp
- Department of Orthopaedics and Rehabilitation, Center for Musculoskeletal Research, University of Rochester Medical Center, 601 Elmwood Ave, Rochester, NY 14642, USA
| | - Joe V Chakkalakal
- Department of Pharmacology and Physiology, University of Rochester Medical Center, 601 Elmwood Ave Box 711, Rochester, NY 14642, USA.,Wilmot Cancer Institute, Stem Cell and Regenerative Medicine Institute, The Rochester Aging Research Center, and Department of Biomedical Engineering, University of Rochester Medical Center, 601 Elmwood Ave, Rochester, NY 14642, USA
| |
Collapse
|
86
|
Mademtzoglou D, Asakura Y, Borok MJ, Alonso-Martin S, Mourikis P, Kodaka Y, Mohan A, Asakura A, Relaix F. Cellular localization of the cell cycle inhibitor Cdkn1c controls growth arrest of adult skeletal muscle stem cells. eLife 2018; 7:33337. [PMID: 30284969 PMCID: PMC6172026 DOI: 10.7554/elife.33337] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2017] [Accepted: 09/02/2018] [Indexed: 01/01/2023] Open
Abstract
Adult skeletal muscle maintenance and regeneration depend on efficient muscle stem cell (MuSC) functions. The mechanisms coordinating cell cycle with activation, renewal, and differentiation of MuSCs remain poorly understood. Here, we investigated how adult MuSCs are regulated by CDKN1c (p57kip2), a cyclin-dependent kinase inhibitor, using mouse molecular genetics. In the absence of CDKN1c, skeletal muscle repair is severely impaired after injury. We show that CDKN1c is not expressed in quiescent MuSCs, while being induced in activated and proliferating myoblasts and maintained in differentiating myogenic cells. In agreement, isolated Cdkn1c-deficient primary myoblasts display differentiation defects and increased proliferation. We further show that the subcellular localization of CDKN1c is dynamic; while CDKN1c is initially localized to the cytoplasm of activated/proliferating myoblasts, progressive nuclear translocation leads to growth arrest during differentiation. We propose that CDKN1c activity is restricted to differentiating myoblasts by regulated cyto-nuclear relocalization, coordinating the balance between proliferation and growth arrest.
Collapse
Affiliation(s)
- Despoina Mademtzoglou
- Inserm, IMRB U955-E10, F-94010, Créteil, France.,Ecole Nationale Veterinaire d'Alfort, Faculté de medecine, F-94000, Université Paris-Est Creteil, Maison Alfort, France
| | - Yoko Asakura
- Stem Cell Institute, Paul and Sheila Wellstone Muscular Dystrophy Center, Department of Neurology, University of Minnesota Medical School, Minneapolis, United States
| | - Matthew J Borok
- Inserm, IMRB U955-E10, F-94010, Créteil, France.,Ecole Nationale Veterinaire d'Alfort, Faculté de medecine, F-94000, Université Paris-Est Creteil, Maison Alfort, France
| | - Sonia Alonso-Martin
- Inserm, IMRB U955-E10, F-94010, Créteil, France.,Ecole Nationale Veterinaire d'Alfort, Faculté de medecine, F-94000, Université Paris-Est Creteil, Maison Alfort, France
| | - Philippos Mourikis
- Inserm, IMRB U955-E10, F-94010, Créteil, France.,Ecole Nationale Veterinaire d'Alfort, Faculté de medecine, F-94000, Université Paris-Est Creteil, Maison Alfort, France
| | - Yusaku Kodaka
- Stem Cell Institute, Paul and Sheila Wellstone Muscular Dystrophy Center, Department of Neurology, University of Minnesota Medical School, Minneapolis, United States
| | - Amrudha Mohan
- Stem Cell Institute, Paul and Sheila Wellstone Muscular Dystrophy Center, Department of Neurology, University of Minnesota Medical School, Minneapolis, United States
| | - Atsushi Asakura
- Stem Cell Institute, Paul and Sheila Wellstone Muscular Dystrophy Center, Department of Neurology, University of Minnesota Medical School, Minneapolis, United States
| | - Frederic Relaix
- Inserm, IMRB U955-E10, F-94010, Créteil, France.,Ecole Nationale Veterinaire d'Alfort, Faculté de medecine, F-94000, Université Paris-Est Creteil, Maison Alfort, France.,Etablissement Français du Sang, Créteil, France.,APHP, Hopitaux Universitaires Henri Mondor, DHU Pepsy & Centre de Référence des Maladies Neuromusculaires GNMH, Créteil, France
| |
Collapse
|
87
|
Fitzpatrick KR, Cucak A, McLoon LK. Changing muscle function with sustained glial derived neurotrophic factor treatment of rabbit extraocular muscle. PLoS One 2018; 13:e0202861. [PMID: 30142211 PMCID: PMC6108505 DOI: 10.1371/journal.pone.0202861] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2018] [Accepted: 08/10/2018] [Indexed: 01/05/2023] Open
Abstract
Recent microarray and RNAseq experiments provided evidence that glial derived neurotrophic factor (GDNF) levels were decreased in extraocular muscles from human strabismic subjects compared to age-matched controls. We assessed the effect of sustained GDNF treatment of the superior rectus muscles of rabbits on their physiological and morphological characteristics, and these were compared to naïve control muscles. Superior rectus muscles of rabbits were implanted with a sustained release pellet of GDNF to deliver 2μg/day, with the contralateral side receiving a placebo pellet. After one month, the muscles were assessed using in vitro physiological methods. The muscles were examined histologically for alteration in fiber size, myosin expression patterns, neuromuscular junction size, and stem cell numbers and compared to age-matched naïve control muscles. GDNF resulted in decreased force generation, which was also seen on the untreated contralateral superior rectus muscles. Muscle relaxation times were increased in the GDNF treated muscles. Myofiber mean cross-sectional areas were increased after the GDNF treatment, but there was a compensatory increase in expression of developmental, neonatal, and slow tonic myosin heavy chain isoforms. In addition, in the GDNF treated muscles there was a large increase in Pitx2-positive myogenic precursor cells. One month of GDNF resulted in significant extraocular muscle adaptation. These changes are interesting relative to the decreased levels of GDNF in the muscles from subjects with strabismus and preliminary data in infant non-human primates where sustained GDNF treatment produced a strabismus. These data support the view that GDNF has the potential for improving eye alignment in subjects with strabismus.
Collapse
Affiliation(s)
- Krysta R. Fitzpatrick
- Department of Ophthalmology and Visual Neurosciences, University of Minnesota, Minneapolis, Minnesota, United States of America
| | - Anja Cucak
- Department of Ophthalmology and Visual Neurosciences, University of Minnesota, Minneapolis, Minnesota, United States of America
| | - Linda K. McLoon
- Department of Ophthalmology and Visual Neurosciences, University of Minnesota, Minneapolis, Minnesota, United States of America
- Department of Ophthalmology and Visual Neurosciences and Department of Neuroscience, University of Minnesota, Minneapolis, Minnesota, United States of America
- * E-mail:
| |
Collapse
|
88
|
Parajuli P, Kumar S, Loumaye A, Singh P, Eragamreddy S, Nguyen TL, Ozkan S, Razzaque MS, Prunier C, Thissen JP, Atfi A. Twist1 Activation in Muscle Progenitor Cells Causes Muscle Loss Akin to Cancer Cachexia. Dev Cell 2018; 45:712-725.e6. [PMID: 29920276 PMCID: PMC6054474 DOI: 10.1016/j.devcel.2018.05.026] [Citation(s) in RCA: 45] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2017] [Revised: 12/11/2017] [Accepted: 05/21/2018] [Indexed: 12/22/2022]
Abstract
Cancer cachexia is characterized by extreme skeletal muscle loss that results in high morbidity and mortality. The incidence of cachexia varies among tumor types, being lowest in sarcomas, whereas 90% of pancreatic ductal adenocarcinoma (PDAC) patients experience severe weight loss. How these tumors trigger muscle depletion is still unfolding. Serendipitously, we found that overexpression of Twist1 in mouse muscle progenitor cells, either constitutively during development or inducibly in adult animals, caused severe muscle atrophy with features reminiscent of cachexia. Using several genetic mouse models of PDAC, we detected a marked increase in Twist1 expression in muscle undergoing cachexia. In cancer patients, elevated levels of Twist1 are associated with greater degrees of muscle wasting. Finally, both genetic and pharmacological inactivation of Twist1 in muscle progenitor cells afforded substantial protection against cancer-mediated cachexia, which translated into meaningful survival benefits, implicating Twist1 as a possible target for attenuating muscle cachexia in cancer patients.
Collapse
Affiliation(s)
- Parash Parajuli
- Cancer Institute, University of Mississippi Medical Center, Jackson, MS 39216, USA
| | - Santosh Kumar
- Cancer Institute, University of Mississippi Medical Center, Jackson, MS 39216, USA
| | - Audrey Loumaye
- Endocrinology, Diabetology, and Nutrition Department, Institut de Recherche Expérimentale et Clinique, Université Catholique de Louvain, Brussels, Belgium
| | - Purba Singh
- Cancer Institute, University of Mississippi Medical Center, Jackson, MS 39216, USA
| | - Sailaja Eragamreddy
- Cancer Institute, University of Mississippi Medical Center, Jackson, MS 39216, USA
| | - Thien Ly Nguyen
- Cancer Institute, University of Mississippi Medical Center, Jackson, MS 39216, USA
| | - Seval Ozkan
- Cancer Institute, University of Mississippi Medical Center, Jackson, MS 39216, USA
| | - Mohammed S Razzaque
- Department of Applied Oral Sciences, The Forsyth Institute, Harvard School of Dental Medicine Affiliate, Cambridge, MA 02142, USA
| | - Céline Prunier
- Sorbonne Universités, UPMC Univ Paris 06, INSERM, Centre de Recherche Saint-Antoine (CRSA), Paris 75012, France
| | - Jean-Paul Thissen
- Endocrinology, Diabetology, and Nutrition Department, Institut de Recherche Expérimentale et Clinique, Université Catholique de Louvain, Brussels, Belgium
| | - Azeddine Atfi
- Cancer Institute, University of Mississippi Medical Center, Jackson, MS 39216, USA; Sorbonne Universités, UPMC Univ Paris 06, INSERM, Centre de Recherche Saint-Antoine (CRSA), Paris 75012, France.
| |
Collapse
|
89
|
Bi P, McAnally JR, Shelton JM, Sánchez-Ortiz E, Bassel-Duby R, Olson EN. Fusogenic micropeptide Myomixer is essential for satellite cell fusion and muscle regeneration. Proc Natl Acad Sci U S A 2018; 115:3864-3869. [PMID: 29581287 PMCID: PMC5899482 DOI: 10.1073/pnas.1800052115] [Citation(s) in RCA: 73] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Regeneration of skeletal muscle in response to injury occurs through fusion of a population of stem cells, known as satellite cells, with injured myofibers. Myomixer, a muscle-specific membrane micropeptide, cooperates with the transmembrane protein Myomaker to regulate embryonic myoblast fusion and muscle formation. To investigate the role of Myomixer in muscle regeneration, we used CRISPR/Cas9-mediated genome editing to generate conditional knockout Myomixer alleles in mice. We show that genetic deletion of Myomixer in satellite cells using a tamoxifen-regulated Cre recombinase transgene under control of the Pax7 promoter abolishes satellite cell fusion and prevents muscle regeneration, resulting in severe muscle degeneration after injury. Satellite cells devoid of Myomixer maintain expression of Myomaker, demonstrating that Myomaker alone is insufficient to drive myoblast fusion. These findings, together with prior studies demonstrating the essentiality of Myomaker for muscle regeneration, highlight the obligatory partnership of Myomixer and Myomaker for myofiber formation throughout embryogenesis and adulthood.
Collapse
Affiliation(s)
- Pengpeng Bi
- Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, TX 75390
- Hamon Center for Regenerative Science and Medicine, University of Texas Southwestern Medical Center, Dallas, TX 75390
- Senator Paul D. Wellstone Muscular Dystrophy Cooperative Research Center, University of Texas Southwestern Medical Center, Dallas, TX 75390
| | - John R McAnally
- Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, TX 75390
- Hamon Center for Regenerative Science and Medicine, University of Texas Southwestern Medical Center, Dallas, TX 75390
- Senator Paul D. Wellstone Muscular Dystrophy Cooperative Research Center, University of Texas Southwestern Medical Center, Dallas, TX 75390
| | - John M Shelton
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX 75390
| | - Efrain Sánchez-Ortiz
- Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, TX 75390
- Hamon Center for Regenerative Science and Medicine, University of Texas Southwestern Medical Center, Dallas, TX 75390
- Senator Paul D. Wellstone Muscular Dystrophy Cooperative Research Center, University of Texas Southwestern Medical Center, Dallas, TX 75390
| | - Rhonda Bassel-Duby
- Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, TX 75390
- Hamon Center for Regenerative Science and Medicine, University of Texas Southwestern Medical Center, Dallas, TX 75390
- Senator Paul D. Wellstone Muscular Dystrophy Cooperative Research Center, University of Texas Southwestern Medical Center, Dallas, TX 75390
| | - Eric N Olson
- Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, TX 75390;
- Hamon Center for Regenerative Science and Medicine, University of Texas Southwestern Medical Center, Dallas, TX 75390
- Senator Paul D. Wellstone Muscular Dystrophy Cooperative Research Center, University of Texas Southwestern Medical Center, Dallas, TX 75390
| |
Collapse
|
90
|
Wang G, Zhu H, Situ C, Han L, Yu Y, Cheung TH, Liu K, Wu Z. p110α of PI3K is necessary and sufficient for quiescence exit in adult muscle satellite cells. EMBO J 2018; 37:embj.201798239. [PMID: 29581096 DOI: 10.15252/embj.201798239] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2017] [Revised: 02/19/2018] [Accepted: 02/23/2018] [Indexed: 11/09/2022] Open
Abstract
Adult mouse muscle satellite cells (MuSCs) are quiescent in uninjured muscles. Upon injury, MuSCs exit quiescence in vivo to become activated, re-enter the cell cycle to proliferate, and differentiate to repair the damaged muscles. It remains unclear which extrinsic cues and intrinsic signaling pathways regulate quiescence exit during MuSC activation. Here, we demonstrated that inducible MuSC-specific deletion of p110α, a catalytic subunit of phosphatidylinositol 3-kinase (PI3K), rendered MuSCs unable to exit quiescence, resulting in severely impaired MuSC proliferation and muscle regeneration. Genetic reactivation of mTORC1, or knockdown of FoxOs, in p110α-null MuSCs partially rescued the above defects, making them key effectors downstream of PI3K in regulating quiescence exit. c-Jun was found to be a key transcriptional target of the PI3K/mTORC1 signaling axis essential for MuSC quiescence exit. Moreover, induction of a constitutively active PI3K in quiescent MuSCs resulted in spontaneous MuSC activation in uninjured muscles and subsequent depletion of the MuSC pool. Thus, PI3K-p110α is both necessary and sufficient for MuSCs to exit quiescence in response to activating signals.
Collapse
Affiliation(s)
- Gang Wang
- Division of Life Science, Center for Stem Cell Research, Center for Systems Biology and Human Health, the State Key Laboratory in Neuroscience, Hong Kong University of Science & Technology, Kowloon Hong Kong, China
| | - Han Zhu
- Division of Life Science, Center for Stem Cell Research, Center for Systems Biology and Human Health, the State Key Laboratory in Neuroscience, Hong Kong University of Science & Technology, Kowloon Hong Kong, China
| | - Chenghao Situ
- Division of Life Science, Center for Stem Cell Research, Center for Systems Biology and Human Health, the State Key Laboratory in Neuroscience, Hong Kong University of Science & Technology, Kowloon Hong Kong, China
| | - Lifang Han
- Division of Life Science, Center for Stem Cell Research, Center for Systems Biology and Human Health, the State Key Laboratory in Neuroscience, Hong Kong University of Science & Technology, Kowloon Hong Kong, China
| | - Youqian Yu
- Division of Life Science, Center for Stem Cell Research, Center for Systems Biology and Human Health, the State Key Laboratory in Neuroscience, Hong Kong University of Science & Technology, Kowloon Hong Kong, China
| | - Tom H Cheung
- Division of Life Science, Center for Stem Cell Research, Center for Systems Biology and Human Health, the State Key Laboratory in Neuroscience, Hong Kong University of Science & Technology, Kowloon Hong Kong, China
| | - Kai Liu
- Division of Life Science, Center for Stem Cell Research, Center for Systems Biology and Human Health, the State Key Laboratory in Neuroscience, Hong Kong University of Science & Technology, Kowloon Hong Kong, China
| | - Zhenguo Wu
- Division of Life Science, Center for Stem Cell Research, Center for Systems Biology and Human Health, the State Key Laboratory in Neuroscience, Hong Kong University of Science & Technology, Kowloon Hong Kong, China
| |
Collapse
|
91
|
Abstract
Down syndrome, caused by trisomy 21, is characterized by a variety of medical conditions including intellectual impairments, cardiovascular defects, blood cell disorders and pre-mature aging phenotypes. Several somatic stem cell populations are dysfunctional in Down syndrome and their deficiencies may contribute to multiple Down syndrome phenotypes. Down syndrome is associated with muscle weakness but skeletal muscle stem cells or satellite cells in Down syndrome have not been investigated. We find that a failure in satellite cell expansion impairs muscle regeneration in the Ts65Dn mouse model of Down syndrome. Ts65Dn satellite cells accumulate DNA damage and over express Usp16, a histone de-ubiquitinating enzyme that regulates the DNA damage response. Impairment of satellite cell function, which further declines as Ts65Dn mice age, underscores stem cell deficiencies as an important contributor to Down syndrome pathologies.
Collapse
|
92
|
Yamamoto M, Legendre NP, Biswas AA, Lawton A, Yamamoto S, Tajbakhsh S, Kardon G, Goldhamer DJ. Loss of MyoD and Myf5 in Skeletal Muscle Stem Cells Results in Altered Myogenic Programming and Failed Regeneration. Stem Cell Reports 2018; 10:956-969. [PMID: 29478898 PMCID: PMC5918368 DOI: 10.1016/j.stemcr.2018.01.027] [Citation(s) in RCA: 103] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2016] [Revised: 01/22/2018] [Accepted: 01/23/2018] [Indexed: 11/22/2022] Open
Abstract
MyoD and Myf5 are fundamental regulators of skeletal muscle lineage determination in the embryo, and their expression is induced in satellite cells following muscle injury. MyoD and Myf5 are also expressed by satellite cell precursors developmentally, although the relative contribution of historical and injury-induced expression to satellite cell function is unknown. We show that satellite cells lacking both MyoD and Myf5 (double knockout [dKO]) are maintained with aging in uninjured muscle. However, injured muscle fails to regenerate and dKO satellite cell progeny accumulate in damaged muscle but do not undergo muscle differentiation. dKO satellite cell progeny continue to express markers of myoblast identity, although their myogenic programming is labile, as demonstrated by dramatic morphological changes and increased propensity for non-myogenic differentiation. These data demonstrate an absolute requirement for either MyoD or Myf5 in muscle regeneration and indicate that their expression after injury stabilizes myogenic identity and confers the capacity for muscle differentiation. MyoD or Myf5 expression in satellite cells is essential for muscle regeneration Satellite cells lacking both regulatory genes exhibit labile myogenic programming A single functional allele of either MyoD or Myf5 can support muscle regeneration Satellite cells lacking both MyoD and Myf5 are maintained with aging
Collapse
Affiliation(s)
- Masakazu Yamamoto
- Department of Molecular & Cell Biology, University of Connecticut Stem Cell Institute, University of Connecticut, 91 N. Eagleville Road, Storrs, CT 06269, USA
| | - Nicholas P Legendre
- Department of Molecular & Cell Biology, University of Connecticut Stem Cell Institute, University of Connecticut, 91 N. Eagleville Road, Storrs, CT 06269, USA
| | - Arpita A Biswas
- Department of Molecular & Cell Biology, University of Connecticut Stem Cell Institute, University of Connecticut, 91 N. Eagleville Road, Storrs, CT 06269, USA
| | - Alexander Lawton
- Department of Molecular & Cell Biology, University of Connecticut Stem Cell Institute, University of Connecticut, 91 N. Eagleville Road, Storrs, CT 06269, USA
| | - Shoko Yamamoto
- Department of Molecular & Cell Biology, University of Connecticut Stem Cell Institute, University of Connecticut, 91 N. Eagleville Road, Storrs, CT 06269, USA
| | - Shahragim Tajbakhsh
- Institut Pasteur, Stem Cells & Development, CNRS URA 2578, 25 rue du Dr. Roux, 75724 Paris Cedex 15, France
| | - Gabrielle Kardon
- Department of Human Genetics, University of Utah, 15 North 2030 East, Salt Lake City, UT 84112, USA
| | - David J Goldhamer
- Department of Molecular & Cell Biology, University of Connecticut Stem Cell Institute, University of Connecticut, 91 N. Eagleville Road, Storrs, CT 06269, USA.
| |
Collapse
|
93
|
Vest KE, Paskavitz AL, Lee JB, Padilla-Benavides T. Dynamic changes in copper homeostasis and post-transcriptional regulation of Atp7a during myogenic differentiation. Metallomics 2018; 10:309-322. [PMID: 29333545 PMCID: PMC5824686 DOI: 10.1039/c7mt00324b] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2017] [Accepted: 01/04/2018] [Indexed: 12/13/2022]
Abstract
Copper (Cu) is an essential metal required for activity of a number of redox active enzymes that participate in critical cellular pathways such as metabolism and cell signaling. Because it is also a toxic metal, Cu must be tightly controlled by a series of transporters and chaperone proteins that regulate Cu homeostasis. The critical nature of Cu is highlighted by the fact that mutations in Cu homeostasis genes cause pathologic conditions such as Menkes and Wilson diseases. While Cu homeostasis in highly affected tissues like the liver and brain is well understood, no study has probed the role of Cu in development of skeletal muscle, another tissue that often shows pathology in these conditions. Here, we found an increase in whole cell Cu content during differentiation of cultured immortalized or primary myoblasts derived from mouse satellite cells. We demonstrate that Cu is required for both proliferation and differentiation of primary myoblasts. We also show that a key Cu homeostasis gene, Atp7a, undergoes dynamic changes in expression during myogenic differentiation. Alternative polyadenylation and stability of Atp7a mRNA fluctuates with differentiation stage of the myoblasts, indicating post-transcriptional regulation of Atp7a that depends on the differentiation state. This is the first report of a requirement for Cu during myogenic differentiation and provides the basis for understanding the network of Cu transport associated with myogenesis.
Collapse
Affiliation(s)
- Katherine E. Vest
- Department of Biology , Emory University , 1510 Clifton Road , Atlanta , GA 30322 , USA
| | - Amanda L. Paskavitz
- Department of Biochemistry and Molecular Pharmacology , University of Massachusetts Medical School , 394 Plantation St. , Worcester , MA 01605 , USA .
| | - Joseph B. Lee
- Department of Biochemistry and Molecular Pharmacology , University of Massachusetts Medical School , 394 Plantation St. , Worcester , MA 01605 , USA .
| | - Teresita Padilla-Benavides
- Department of Biochemistry and Molecular Pharmacology , University of Massachusetts Medical School , 394 Plantation St. , Worcester , MA 01605 , USA .
| |
Collapse
|
94
|
Kneppers A, Verdijk L, de Theije C, Corten M, Gielen E, van Loon L, Schols A, Langen R. A novel in vitro model for the assessment of postnatal myonuclear accretion. Skelet Muscle 2018; 8:4. [PMID: 29444710 PMCID: PMC5813369 DOI: 10.1186/s13395-018-0151-4] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2017] [Accepted: 01/26/2018] [Indexed: 01/21/2023] Open
Abstract
BACKGROUND Due to the post-mitotic nature of myonuclei, postnatal myogenesis is essential for skeletal muscle growth, repair, and regeneration. This process is facilitated by satellite cells through proliferation, differentiation, and subsequent fusion with a pre-existing muscle fiber (i.e., myonuclear accretion). Current knowledge of myogenesis is primarily based on the in vitro formation of syncytia from myoblasts, which represents aspects of developmental myogenesis, but may incompletely portray postnatal myogenesis. Therefore, we aimed to develop an in vitro model that better reflects postnatal myogenesis, to study the cell intrinsic and extrinsic processes and signaling involved in the regulation of postnatal myogenesis. METHODS Proliferating C2C12 myoblasts were trypsinized and co-cultured for 3 days with 5 days differentiated C2C12 myotubes. Postnatal myonuclear accretion was visually assessed by live cell time-lapse imaging and cell tracing by cell labeling with Vybrant® DiD and DiO. Furthermore, a Cre/LoxP-based cell system was developed to semi-quantitatively assess in vitro postnatal myonuclear accretion by the conditional expression of luciferase upon myoblast-myotube fusion. Luciferase activity was assessed luminometrically and corrected for total protein content. RESULTS Live cell time-lapse imaging, staining-based cell tracing, and recombination-dependent luciferase activity, showed the occurrence of postnatal myonuclear accretion in vitro. Treatment of co-cultures with the myogenic factor IGF-I (p < 0.001) and the cytokines IL-13 (p < 0.05) and IL-4 (p < 0.001) increased postnatal myonuclear accretion, while the myogenic inhibitors cytochalasin D (p < 0.001), myostatin (p < 0.05), and TNFα (p < 0.001) decreased postnatal myonuclear accretion. Furthermore, postnatal myonuclear accretion was increased upon recovery from electrical pulse stimulation-induced fiber damage (p < 0.001) and LY29004-induced atrophy (p < 0.001). Moreover, cell type-specific siRNA-mediated knockdown of myomaker in myoblasts (p < 0.001), but not in myotubes, decreased postnatal myonuclear accretion. CONCLUSIONS We developed a physiologically relevant, sensitive, high-throughput cell system for semi-quantitative assessment of in vitro postnatal myonuclear accretion, which can be used to mimic physiological myogenesis triggers, and can distinguish the cell type-specific roles of signals and responses in the regulation of postnatal myogenesis. As such, this method is suitable for both basal and translational research on the regulation of postnatal myogenesis, and will improve our understanding of muscle pathologies that result from impaired satellite cell number or function.
Collapse
Affiliation(s)
- Anita Kneppers
- Department of Respiratory Medicine, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Centre+, Maastricht, The Netherlands.
| | - Lex Verdijk
- Department of Human Biology and Movement Sciences, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Centre+, Maastricht, The Netherlands
| | - Chiel de Theije
- Department of Respiratory Medicine, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Centre+, Maastricht, The Netherlands
| | - Mark Corten
- Department of Respiratory Medicine, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Centre+, Maastricht, The Netherlands
| | - Ellis Gielen
- Department of Respiratory Medicine, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Centre+, Maastricht, The Netherlands
| | - Luc van Loon
- Department of Human Biology and Movement Sciences, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Centre+, Maastricht, The Netherlands
| | - Annemie Schols
- Department of Respiratory Medicine, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Centre+, Maastricht, The Netherlands
| | - Ramon Langen
- Department of Respiratory Medicine, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Centre+, Maastricht, The Netherlands
| |
Collapse
|
95
|
Gadek KE, Wang H, Hall MN, Sungello M, Libby A, MacLaskey D, Eckel RH, Olwin BB. Striated muscle gene therapy for the treatment of lipoprotein lipase deficiency. PLoS One 2018; 13:e0190963. [PMID: 29304082 PMCID: PMC5755938 DOI: 10.1371/journal.pone.0190963] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2017] [Accepted: 12/22/2017] [Indexed: 11/19/2022] Open
Abstract
Excessive circulating triglycerides due to reduction or loss of lipoprotein lipase activity contribute to hypertriglyceridemia and increased risk for pancreatitis. The only gene therapy treatment for lipoprotein lipase deficiency decreases pancreatitis but minimally reduces hypertriglyceridemia. Synthesized in multiple tissues including striated muscle and adipose tissue, lipoprotein lipase is trafficked to blood vessel endothelial cells where it is anchored at the plasma membrane and hydrolyzes triglycerides into free fatty acids. We conditionally knocked out lipoprotein lipase in differentiated striated muscle tissue lowering striated muscle lipoprotein lipase activity causing hypertriglyceridemia. We then crossed lipoprotein lipase striated muscle knockout mice with mice possessing a conditional avian retroviral receptor gene and injected mice with either a human lipoprotein lipase retrovirus or an mCherry control retrovirus. Post-heparin plasma lipoprotein lipase activity increased for three weeks following human lipoprotein lipase retroviral infection compared to mCherry infected mice. Human lipoprotein lipase infected mice had significantly lower blood triglycerides compared to mCherry controls and were comparable to wild-type blood triglyceride levels. Thus, targeted delivery of human lipoprotein lipase into striated muscle tissue identifies a potential therapeutic target for lipoprotein lipase deficiency.
Collapse
Affiliation(s)
- Katherine E. Gadek
- Department of Molecular, Cellular and Developmental Biology, University of Colorado Boulder, Boulder, Colorado United States of America
| | - Hong Wang
- Division of Endocrinology, Metabolism, and Diabetes, University of Colorado Denver Anschutz Medical Campus, Aurora, Colorado United States of America
| | - Monica N. Hall
- Department of Molecular, Cellular and Developmental Biology, University of Colorado Boulder, Boulder, Colorado United States of America
| | - Mitchell Sungello
- Division of Endocrinology, Metabolism, and Diabetes, University of Colorado Denver Anschutz Medical Campus, Aurora, Colorado United States of America
| | - Andrew Libby
- Division of Endocrinology, Metabolism, and Diabetes, University of Colorado Denver Anschutz Medical Campus, Aurora, Colorado United States of America
| | - Drew MacLaskey
- Department of Molecular, Cellular and Developmental Biology, University of Colorado Boulder, Boulder, Colorado United States of America
| | - Robert H. Eckel
- Division of Endocrinology, Metabolism, and Diabetes, University of Colorado Denver Anschutz Medical Campus, Aurora, Colorado United States of America
- * E-mail: (BBO); (RHE)
| | - Bradley B. Olwin
- Department of Molecular, Cellular and Developmental Biology, University of Colorado Boulder, Boulder, Colorado United States of America
- * E-mail: (BBO); (RHE)
| |
Collapse
|
96
|
Tierney MT, Stec MJ, Rulands S, Simons BD, Sacco A. Muscle Stem Cells Exhibit Distinct Clonal Dynamics in Response to Tissue Repair and Homeostatic Aging. Cell Stem Cell 2018; 22:119-127.e3. [PMID: 29249462 PMCID: PMC5945549 DOI: 10.1016/j.stem.2017.11.009] [Citation(s) in RCA: 57] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2017] [Revised: 09/15/2017] [Accepted: 11/07/2017] [Indexed: 10/18/2022]
Abstract
The clonal complexity of adult stem cell pools is progressively lost during homeostatic turnover in several tissues, suggesting a decrease in the number of stem cells with distinct clonal origins. The functional impact of reduced complexity on stem cell pools, and how different tissue microenvironments may contribute to such a reduction, are poorly understood. Here, we performed clonal multicolor lineage tracing of skeletal muscle stem cells (MuSCs) to address these questions. We found that MuSC clonal complexity is maintained during aging despite heterogenous reductions in proliferative capacity, allowing aged muscle to mount a clonally diverse, albeit diminished, response to injury. In contrast, repeated bouts of tissue repair cause a progressive reduction in MuSC clonal complexity indicative of neutral drift. Consistently, biostatistical modeling suggests that MuSCs undergo symmetric expansions with stochastic fate acquisition during tissue repair. These findings establish distinct principles that underlie stem cell dynamics during homeostatic aging and muscle regeneration.
Collapse
Affiliation(s)
- Matthew T Tierney
- Graduate School of Biomedical Sciences, Sanford Burnham Prebys Medical Discovery Institute, 10901 N. Torrey Pines Road, La Jolla, CA 92037, USA; Development, Aging and Regeneration Program, Sanford Burnham Prebys Medical Discovery Institute, 10901 N. Torrey Pines Road, La Jolla, CA 92037, USA
| | - Michael J Stec
- Development, Aging and Regeneration Program, Sanford Burnham Prebys Medical Discovery Institute, 10901 N. Torrey Pines Road, La Jolla, CA 92037, USA
| | - Steffen Rulands
- Cavendish Laboratory, Department of Physics, J. J. Thomson Avenue, Cambridge CB3 0HE, UK; The Wellcome Trust/Cancer Research UK Gurdon Institute and Wellcome Trust-Medical Research Council Stem Cell Institute, University of Cambridge, Tennis Court Road, Cambridge CB2 1QN, UK
| | - Benjamin D Simons
- Cavendish Laboratory, Department of Physics, J. J. Thomson Avenue, Cambridge CB3 0HE, UK; The Wellcome Trust/Cancer Research UK Gurdon Institute and Wellcome Trust-Medical Research Council Stem Cell Institute, University of Cambridge, Tennis Court Road, Cambridge CB2 1QN, UK
| | - Alessandra Sacco
- Development, Aging and Regeneration Program, Sanford Burnham Prebys Medical Discovery Institute, 10901 N. Torrey Pines Road, La Jolla, CA 92037, USA.
| |
Collapse
|
97
|
Murach KA, Fry CS, Kirby TJ, Jackson JR, Lee JD, White SH, Dupont-Versteegden EE, McCarthy JJ, Peterson CA. Starring or Supporting Role? Satellite Cells and Skeletal Muscle Fiber Size Regulation. Physiology (Bethesda) 2018; 33:26-38. [PMID: 29212890 PMCID: PMC5866409 DOI: 10.1152/physiol.00019.2017] [Citation(s) in RCA: 79] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2017] [Revised: 09/18/2017] [Accepted: 09/18/2017] [Indexed: 12/22/2022] Open
Abstract
Recent loss-of-function studies show that satellite cell depletion does not promote sarcopenia or unloading-induced atrophy, and does not prevent regrowth. Although overload-induced muscle fiber hypertrophy is normally associated with satellite cell-mediated myonuclear accretion, hypertrophic adaptation proceeds in the absence of satellite cells in fully grown adult mice, but not in young growing mice. Emerging evidence also indicates that satellite cells play an important role in remodeling the extracellular matrix during hypertrophy.
Collapse
Affiliation(s)
- Kevin A Murach
- The Center for Muscle Biology, University of Kentucky, Lexington, Kentucky
- Department of Rehabilitation Sciences, College of Health Sciences, University of Kentucky, Lexington, Kentucky
| | - Christopher S Fry
- Department of Nutrition and Metabolism, School of Health Professions, University of Texas Medical Branch at Galveston, Galveston, Texas
| | - Tyler J Kirby
- The Weill Institute for Cell and Molecular Biology, Cornell University, Ithaca, New York
| | - Janna R Jackson
- The Center for Muscle Biology, University of Kentucky, Lexington, Kentucky
- Department of Rehabilitation Sciences, College of Health Sciences, University of Kentucky, Lexington, Kentucky
| | - Jonah D Lee
- Environment, Health, and Safety, University of Michigan, Ann Arbor, Michigan
| | - Sarah H White
- Department of Animal Science, Texas A&M University, College Station, Texas; and
| | - Esther E Dupont-Versteegden
- The Center for Muscle Biology, University of Kentucky, Lexington, Kentucky
- Department of Rehabilitation Sciences, College of Health Sciences, University of Kentucky, Lexington, Kentucky
| | - John J McCarthy
- The Center for Muscle Biology, University of Kentucky, Lexington, Kentucky
- Department of Physiology, College of Medicine, University of Kentucky, Lexington, Kentucky
| | - Charlotte A Peterson
- The Center for Muscle Biology, University of Kentucky, Lexington, Kentucky;
- Department of Rehabilitation Sciences, College of Health Sciences, University of Kentucky, Lexington, Kentucky
| |
Collapse
|
98
|
Klose A, Liu W, Paris ND, Forman S, Krolewski JJ, Nastiuk KL, Chakkalakal JV. Castration induces satellite cell activation that contributes to skeletal muscle maintenance. JCSM RAPID COMMUNICATIONS 2018; 1:e00040. [PMID: 29782610 PMCID: PMC5959044] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Subscribe] [Scholar Register] [Indexed: 06/08/2023]
Abstract
BACKGROUND Sarcopenia, the age-related loss of skeletal muscle, is a side effect of androgen deprivation therapy (ADT) for prostate cancer patients. Resident stem cells of skeletal muscle, satellite cells (SCs), are an essential source of progenitors for the growth and regeneration of skeletal muscle. Decreased androgen signaling and deficits in the number and function of SCs are features of aging. Although androgen signaling is known to regulate skeletal muscle, the cellular basis for ADT-induced exacerbation of sarcopenia is unknown. Furthermore, the consequences of androgen deprivation on SC fate in adult skeletal muscle remain largely unexplored. METHODS We examined SC fate in an androgen-deprived environment using immunofluorescence and fluorescence-activated cell sorting (FACS) with SC-specific markers in young castrated mice. To study the effects of androgen deprivation on SC function and skeletal muscle regenerative capacity, young castrated mice were subjected to experimental regenerative paradigms. SC-derived-cell contributions to skeletal muscle maintenance were examined in castrated Pax7CreER/+; ROSA26mTmG/+ mice. SCs were depleted in Pax7CreER/+; ROSA26DTA/+ mice to ascertain the consequences of SC ablation in sham and castrated skeletal muscles. Confocal immunofluorescence analysis of neuromuscular junctions (NMJs), and assessment of skeletal muscle physiology, contractile properties, and integrity were conducted. RESULTS Castration led to SC activation, however this did not result in a decline in SC function or skeletal muscle regenerative capacity. Surprisingly, castration induced SC-dependent maintenance of young skeletal muscle. The functional dependence of skeletal muscles on SCs in young castrated mice was demonstrated by an increase in SC-derived-cell fusion within skeletal muscle fibers. SC depletion was associated with further atrophy and functional decline, as well as the induction of partial innervation and the loss of NMJ-associated myonuclei in skeletal muscles from castrated mice. CONCLUSION The maintenance of skeletal muscles in young castrated mice relies on the cellular contributions of SCs. Considering the well-described age-related decline in SCs, the results in this study highlight the need to devise strategies that promote SC maintenance and activity to attenuate or reverse the progression of sarcopenia in elderly androgen-deprived individuals.
Collapse
Affiliation(s)
- Alanna Klose
- Department of Orthopaedics and Rehabilitation, Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, NY USA
| | - Wenxuan Liu
- Department of Orthopaedics and Rehabilitation, Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, NY USA
| | - Nicole D. Paris
- Department of Orthopaedics and Rehabilitation, Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, NY USA
| | - Sophie Forman
- Department of Orthopaedics and Rehabilitation, Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, NY USA
| | - John J. Krolewski
- Department of Cancer Genetics & Genomics, and Center for Personalized Medicine, Roswell Park Cancer Institute; Buffalo, NY USA
| | - Kent L. Nastiuk
- Department of Cancer Genetics & Genomics, and Department of Urology, Roswell Park Cancer Institute; Buffalo, NY USA
| | - Joe V. Chakkalakal
- Department of Orthopaedics and Rehabilitation, Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, NY USA
- Stem Cell and Regenerative Medicine Institute, University of Rochester Medical Center, Rochester, NY USA
- The Rochester Aging Research Center, University of Rochester Medical Center, Rochester, NY USA
| |
Collapse
|
99
|
Dyar KA, Schiaffino S, Blaauw B. Inactivation of the intrinsic muscle clock does not cause sarcopenia. J Physiol 2017; 594:3161-2. [PMID: 27246551 DOI: 10.1113/jp272127] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2016] [Revised: 01/13/2016] [Indexed: 01/09/2023] Open
Affiliation(s)
- K A Dyar
- Venetian Institute of Molecular Medicine (VIMM), 35129, Padova, Italy
| | - S Schiaffino
- Venetian Institute of Molecular Medicine (VIMM), 35129, Padova, Italy
| | - B Blaauw
- Venetian Institute of Molecular Medicine (VIMM), 35129, Padova, Italy.
| |
Collapse
|
100
|
Hebert SL, Fitzpatrick KR, McConnell SA, Cucak A, Yuan C, McLoon LK. Effects of retinoic acid signaling on extraocular muscle myogenic precursor cells in vitro. Exp Cell Res 2017; 361:101-111. [PMID: 29017757 PMCID: PMC6546114 DOI: 10.1016/j.yexcr.2017.10.007] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2017] [Revised: 10/03/2017] [Accepted: 10/05/2017] [Indexed: 02/04/2023]
Abstract
One major difference between limb and extraocular muscles (EOM) is the presence of an enriched population of Pitx2-positive myogenic precursor cells in EOM compared to limb muscle. We hypothesize that retinoic acid regulates Pitx2 expression in EOM myogenic precursor cells and that its effects would differ in leg muscle. The two muscle groups expressed differential retinoic acid receptor (RAR) and retinoid X receptor (RXR) levels. RXR co-localized with the Pitx2-positive cells but not with those expressing Pax7. EOM-derived and LEG-derived EECD34 cells were treated with vehicle, retinoic acid, the RXR agonist bexarotene, the RAR inverse agonist BMS493, or the RXR antagonist UVI 3003. In vitro, fewer EOM-derived EECD34 cells expressed desmin and fused, while more LEG-derived cells expressed desmin and fused when treated with retinoic acid compared to vehicle. Both EOM and LEG-derived EECD34 cells exposed to retinoic acid showed a higher percentage of cells expressing Pitx2 compared to vehicle, supporting the hypothesis that retinoic acid plays a role in maintaining Pitx2 expression. We hypothesize that retinoic acid signaling aids in the maintenance of large numbers of undifferentiated myogenic precursor cells in the EOM, which would be required to maintain EOM normalcy throughout a lifetime of myonuclear turnover.
Collapse
Affiliation(s)
- Sadie L Hebert
- Department of Ophthalmology and Visual Neurosciences, University of Minnesota, Minneapolis, MN, United States
| | - Krysta R Fitzpatrick
- Department of Ophthalmology and Visual Neurosciences, University of Minnesota, Minneapolis, MN, United States
| | - Samantha A McConnell
- Department of Ophthalmology and Visual Neurosciences, University of Minnesota, Minneapolis, MN, United States
| | - Anja Cucak
- Department of Ophthalmology and Visual Neurosciences, University of Minnesota, Minneapolis, MN, United States
| | - Ching Yuan
- Department of Ophthalmology and Visual Neurosciences, University of Minnesota, Minneapolis, MN, United States
| | - Linda K McLoon
- Department of Ophthalmology and Visual Neurosciences, University of Minnesota, Minneapolis, MN, United States; Department of Neuroscience, University of Minnesota, Minneapolis, MN, United States.
| |
Collapse
|